EP3898685A1 - Clec12axcd3 bispecific antibodies and methods for the treatment of disease - Google Patents

Clec12axcd3 bispecific antibodies and methods for the treatment of disease

Info

Publication number
EP3898685A1
EP3898685A1 EP19835540.6A EP19835540A EP3898685A1 EP 3898685 A1 EP3898685 A1 EP 3898685A1 EP 19835540 A EP19835540 A EP 19835540A EP 3898685 A1 EP3898685 A1 EP 3898685A1
Authority
EP
European Patent Office
Prior art keywords
cells
clec12a
antibody
bispecific antibody
day
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19835540.6A
Other languages
German (de)
English (en)
French (fr)
Inventor
Alexander Berthold Hendrik Bakker
Cornelis Jacob Johannes George BOL
Pieter Fokko VAN LOO
Leonardo Andres SIRULNIK
Ernesto Isaac WASSERMAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merus BV
Original Assignee
Merus BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merus BV filed Critical Merus BV
Publication of EP3898685A1 publication Critical patent/EP3898685A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the invention relates to the field of antibodies, in particular to the field of therapeutic antibodies.
  • the antibodies can be used in the treatment of humane.
  • the invention relates to antibodies and preferably bispecific antibodies for the treatment of a cancer.
  • AML Acute myeloid leukaemia
  • the present invention focuses on an antigen specifically expressed on AML blasts and leukaemic stem cells (LSCs), which is the C-type lectin domain family 12 member A or CLEC12A (also referred to as human myeloid inhibitory C-type lectin, hMICL, C-type lectin-like molecule- 1, CLL-1, or CD371) (Bakker, van den Oudenrijn et al. 2004, van Bhenen, van Dongen et al. 2007).
  • CLEC12A is a myeloid differentiation antigen that is expressed on leukemic cells in 90-95% of cases of either de novo or relapsed AML, irrespective of subtype (Bakker, van den
  • CLEC12A is selectively expressed on CD34POSCD38NEG LSCs but, based on in vitro and vivo evidence, it is not expressed on normal hematopoietic stem cells (HSCs), erythroid precursors or megakaryocytes (van Rhenen, van Dongen et al. 2007, Kikushige and Miyamoto 2013).
  • HSCs normal hematopoietic stem cells
  • erythroid precursors or megakaryocytes
  • CLECl2AxCD3 bispecific antibodies induce efficient T cell- mediated CLEC12A targeting and selectively eradicates leukaemic cells (including leukaemic stem cells) without affecting normal HSCs, thereby permitting normal haematopoiesis to be re-established swiftly after treatment and thereby limiting haematological toxicity.
  • Many of the T cell engager formats face issues, including a short serum half- life, immunogenicity and difficulties in terms of manufacture (Brinkmann and Kontermann 2017).
  • the CLECl2AxCD3 bispecific antibodies of the present invention have a good serum half-life. The means and methods of the invention do not significantly induce an immune response against the therapeutic agent.
  • the invention provides a method of treating a subject for a CLEC12A positive cancer the method comprising treating the subject in need thereof with two or more administrations of a bispecific antibody that binds CDS and CLEG 12A, wherein in a first administration a first amount of the bispecific antibody is administered and wherein in each of the subsequent administrations the amount of bispecific antibody is higher than the amount of bispecific antibody in the first administration.
  • the invention also provides a bispecific antibody that binds CDS and CLEC12A for use in a method of treatment of CLEC12A positive cancer in a subject, wherein said treatment comprises two or more administrations of a bispecific antibody that binds CDS and CLEC12A, wherein in a first
  • a first amount of the bispecific antibody is administered and wherein in each of the subsequent administrations the amount of bispecific antibody is higher than the amount of bispecific antibody in the first
  • the invention provides a method of purging CLEC12A positive hemopoietic cells, preferably CLEC12A positive malignant cells, from a subject and repopulating the hemopoietic system of said subject with normal cells, the method comprising administering to the subject in need thereof a bispecific antibody that binds CD3 and CLEC12A at intervals thereby killing CLEC12A positive malignant cells, and stimulating hemopoietic stem cells and/or hemopoietic progenitor cells of said subject to repopulate said hemopoietic system with newly formed hemopoietic stem cell derived cells, including CLEC12A positive cells.
  • the hematopoietic stem cells reside in the bone marrow.
  • the presence of deleterious CLEC12A positive cells can deprive healthy cells of nutrients and occupy the compartment thereby limiting normal growth and development of granulocytes, macrophage stem cells, which themselves give rise to basophil, neutrophil, eosinophil and monocyte blood cells.
  • replenishment of healthy hemopoietic stem cells is stimulated by increasing availability of nutrients and space in the bone marrow compartment for their growth, survival and differentiation into downstream CLEC12A positive and CLEC12A negative, healthy cells, such as basophil, neutrophil, eosinophil and monocyte blood cells.
  • the subject is preferably a subject that is diagnosed to have acute myeloid leukaemia (AML), myelodysplastic syndrome (MDS), myelofibrosis or
  • myeloproliferative neoplasm blast phase MPN-BF.
  • a method of preparing an autologous bone marrow cell graft for a subject that is being treated for a CLEC12A positive cancer comprising incubating a bone marrow cell preparation of said subject with a bispecific antibody that binds CDS and CLEC12A under conditions suitable for killing of CLEC12A positive cells and subsequently collecting bone marrow cells from said incubation.
  • the invention further provides a method of treating a CLEC12A positive cancer in a subject, the method comprising incubating a bone marrow cell preparation of said subject with a bispecific antibody that binds CDS and
  • CLEC12A under conditions suitable for killing of CLEC12A positive cells, treating the subject with an hemopoietic system ablative therapy and providing said subject with a bone marrow cell graft comprising said incubated bone marrow cells.
  • a method of providing a subject with a hemopoietic stem cell sparing cancer treatment comprising administering a bispecific antibody that binds CDS and CLEC12A to the subject in need thereof.
  • the subject preferably has a CLEC12A positive cancer.
  • CLECl2AxCD3 bispecific antibodies specifically bind to CLECl2A pos and CDS 1*08 target cells.
  • A Binding of a CLECl2AxCD3 bispecific antibody to CLEC12A and CDS was determined on a panel of tumour cell lines by flow cytometry.
  • HL60 CLECl2A POS promyelogenous cell line
  • CLEC12A-CHO-K1 CHO-K1 cell line stably expressing human CLEC12A (validated with reference anti-CLEC12A antibody, data not shown)
  • CHO-K1 CLEC12A NEG parental CHO- K1 cell line
  • Jurkat E6.1 CDS 1*08 T cell line
  • J.RT-T3.5 CDS ⁇ 0 T cell line.
  • B A CLECl2AxCD3 bispecific antibody binding profile in normal peripheral blood by flow cytometry.
  • CD4 T cells CD4 1*08 ; CDS T cells: CD8TM 8 ; NK cells: CD3 li®G CD56 TOS ; NKT cells: CDS ⁇ CD56 POS B cells: CD19 R3 ⁇ 45 ; myeloid dendritic cells (DC): BDCA 1 POS CD 19 N m , granulocytes: based on SSC-FSC; monocytes: CD14 TOii CD33 pos .
  • C A CLEC12AxCD3 bispecific antibody binding within CD34 P0S progenitor compartment in normal bone marrow by flow cytometry.
  • HSC hemopoietic stem cell
  • MPP multipotent progenitor
  • CMP multipotent progenitor
  • CLEC12A pos HL60 cells Resting T cells derived from healthy donors were purified by negative selection and co -cultured with CFSE-labelled HL60 cells in the presence of antibody MF4327xMF5196, MockxCDS, control IgG or PBS.
  • A-B Cells were co-incubated at an E:T ratio of 5:1 together with indicated IgGs - all with WT Fc effector function - at an IgG concentration of 1000 ng/mL.
  • C-E Cells were co-incubated at an E:T ratio of 6:1 with indicated IgGs— all with silenced Fc effector function - at a range of IgG concentrations.
  • the capacity of antibody MF4327xMF5196 (with silenced Fc effector function) to induce activation of CD4 and CDS T cells (C, D) and target cell lysis (E) was quantified after 48 hours by flow cytometry relative to the PBS condition.
  • the data shown are from a representative donor, whereby a total of 6 donors were tested in 3 independent assays.
  • antibody MF4327xMF5196 induces CLECl2A-mediated lysis of monocytes, and inflammatory cytokine release, and has the capacity to induce CLEC12A-antigen specific T cell proliferation.
  • A-C Healthy donor-derived PBMC samples were co-cultured with antibody MF4327xMF5196 or MockxCD3 antibody at the indicated concentration range and compared to the PBS condition. T cell activation (A), monocyte lysis by autologous T cells (B) and B cell lysis (C) were assessed after 48 hours using flow cytometry.
  • cytokine levels were detectable for the six cytokines shown.
  • Data shown in A-D are from a single donor and are representative of 4 independent donors from multiple independent experiments.
  • Data shown in E are from two representative healthy PBMC donors, with duplicates for each donor, whereby PBMCs from 8 healthy donors were tested.
  • antibody MF4327xMF5196 spares the potential of normal bone marrow progenitor cells to develop the complete myeloid and erythroid lineage.
  • CD34 pos hemopoietic progenitor cells from healthy donor bone marrow were co -cultured with autologous pre- activated T cells in the presence of 200 ng/mL antibody MF4327xMF5196 or MockxCDS at an E:T ratio of 10:1 for 16 hours.
  • A) Using a quantified flow cytometry setup, antibody MF4327xMF5196- induced lysis was determined relative to non- IgG condition for the indicated cell fractions. Each data point represents the mean value for an independent bone marrow donor tested in triplicate (n 4).
  • CD34 FOS cells Capacity of CD34 FOS cells upon 16 hours cytotoxicity assay with antibody MF4327xMF5196 to give rise to granulocyte- macrophage colonies (CFU-GM), burst-forming unite-erythrocyte (BFU-E) and megakaryocyte colonies (CFU-Mk) was quantified after two weeks culture in semi- solid medium, from four different donors. Control cultures are either non-IgG treated co-cultures or CD34 POS cells seeded alone. Each data point represents the mean value of triplicate cultures
  • CFU-GM and BFU-E or quadruplicates (CFU-Mk) per donor.
  • E Flow cytometric analysis of CD15rosCD33 ⁇ s and CD 14 P0S CD36 POS CD33 P0S cell content of the two week MethoCult cultures described in D. Data show the frequency of monocytic and myelocytic cells out of total CD33 P0S cells for three different donors.
  • HSC hemopoietic stem cell
  • MPP multipotent progenitor
  • CMP common myeloid progenitor
  • GMP granulocyte-macrophage progenitor
  • MEP megakaryocyte - erythroid progenitor
  • CLP common lymphoid progenitor.
  • Statistical analysis was performed using paired student's t test. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
  • CDS POS T cells isolated from the peripheral blood of AML patients in clinical remission were co-cultured for 48 hours with CFSE-labelled CLEC12A P0S autologous primary AML blasts collected at AML diagnosis.
  • the E:T ratio was 5:1 and IgGs were added at a concentration of 1,000 ng/mL: antibody
  • AML blast lysis induced by each IgG relative to the PBS condition was quantified using flow cytometry.
  • A Diagram showing stage of AML at which the AML blast samples and autologous T cells were obtained.
  • B Expression of CLEC12A antigen on primary AML blast samples (AML blast defined as 0045*
  • the histograms show anti-CLECl2A IgG binding for each patient sample, with lymphocytes indicated in grey and AML blasts in black.
  • CLEC12A positivity was set based on the CLECl2A-negati.ve lymphocyte fraction; percentage CLEC12A positive blasts is given.
  • C-D Activation status of AML patient CD4 pos (C) and CD8 PfJS T cells (D).
  • Fig 7. Gating strategy used to quantify HL60 target cell lysis by flow cytometry. A fixed volume of cells was measured by flow cytometry. Live cells were gated based on the FSC-SSC profile (left-hand plots). Within the live gate, the CFSE 1*05 HL60 cells were then gated (right-hand plots). Absolute numbers of CFSE pos live HL60 target cells within that gate were used to calculate the percentage of HL60 target cell lysis as described in the materials and methods section. Upper plots show a PBS condition with no cytotoxicity, while lower plots show an antibody MF4327xMF5196 condition in which HL60 cytotoxicity was observed.
  • Fig 8. Gating strategy used to quantify cytotoxicity in PBMC-based monocyte cytotoxicity assay.
  • Monocytes CD14 P0B
  • CD4 CD4 pos CD3 p08
  • CDS CDS ⁇ CDSTM 8
  • B cells CD 19 ⁇
  • the absolute number of monocytes and B cells measured in a fixed volume of assay medium was quantified and used for calculating the percentage of cytotoxicity induced by the test IgG condition (lower panel) relative to the PBS control condition (upper panel) as described in the materials and methods section.
  • CD4 pos and CDS 1*08 T cell subsets were identified within the T cell population, and their activation status was determined using the activation marker CD69, relative to the PBS control condition.
  • Fig. 10 Gating strategy used to quantify recovery of healthy donor bone marrow CD34 poe progenitor cells.
  • CD34 P0S cells were gated from the co-cultured T cells using CD34 and CD5.
  • CD34 POS cells Within total CD34 POS cells, cells were divided into three populations based on CLEC12A and CD 10 expression. For quantification of the CD34 P0S populations, a fixed volume of flow count fhiorospheres of known concentration was added to each sample before acquisition. The absolute number of cells per mL was calculated based on the ratio of fluorospheres and cells.
  • Fig. 11 Gating strategy used to determine identity and relative ratio of different CD34 POS progenitor cells. First single cells were gated based on FSC and SSC area and width, and then live cells were defined based on FSC- SSC profile and 7AAD negativity. Within live cells, lineage negative CD34 POS cells were gated, followed by a FSC-SSC back gate. Cells were subsequently divided into CD38 pos and CD38 NKG compartments. Within the CD34 pos CD38 pos compartment, CLPs were gated on based on CD 10 expression. Cells lacking CD 10 expression were further separated into CMP, MEP and GMP based on CD123 and CD45RA expression. For some donors, CD 135 was used instead of CD123 for defining these three populations.
  • HSC hemopoietic stem cell
  • MPP multipotent progenitor
  • CMP common myeloid progenitor
  • GMP granulocyte-macrophage progenitor
  • MEP megakaryocyte- erythroid progenitor
  • CLP common lymphoid progenitor.
  • antibody MF4327xMF5196 redirects AML patient T cells and healthy donor T cells to specifically lyse CLEC12A pos AML cell line with equal efficiency.
  • Resting CD3 POS T cells were isolated from the peripheral blood of six healthy donors or six AML patients in clinical remission (patients 1-6; see Table III for patient characteristics). Isolated T cells were co-cultured with CFSE- labelled HL60 target cells at an E:T ratio of 6:1 for 48 hours in the presence of 1,000 ng/mL antibody MF4327xMF5196, MockxCD3 IgG or control IgG (all with WT Fc effector function).
  • A-B Flow cytometry to analyse the subset composition of the CD4 pos T cells (A) and CDe* 08 T cells (B) within the 003 ⁇ fraction of T cells taken from healthy donors (HD) or AML patients subsequently used for HL60 target cell lysis.
  • TCM central memory T cells
  • T NAIVE naive T cells
  • TKMKL+ effector- memory cells with reacquired CD45RA
  • TEM effector memory T cells.
  • CD4 POS T cells C
  • CD8TM 8 T cells D
  • HL60 target cell lysis E
  • FIG. 13 Morphology and JAK2V617F Mutational Status of Cells Generated in Secondary Plating of SSC ,ow CD45 dhn CD34 + CD38CLEC12A* / Cells. May-Giemsa staining of re-plated CLEC12A positive cells, individual colonies were picked and tested for the JAK2 driver mutation.
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 15 mg on day 8, and a full dose of 26 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 16 mg on day 8, and a full dose of 25 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 15 mg on day 8, and a full dose of 25 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 16 mg on day 8, and a full dose of 25 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15. Cytokine levels were also determined at the end of a treatment cyde (day 28). Figure 15b. IL-8 levels of patients treated with antibody
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28). Figure 16d. IFNy levels of patient treated with antibody
  • MF4327xMF5196 In PG/ml, measured before dosing, 4 hours and 24 hours after the end of infusion. Dosing took place on day 1, day 4, day 8, and day 22. The patients received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15. Cytokine levels were also determined at the end of a treatment cycle (day 28).
  • Cytokine release syndrome is a form of systemic inflammatory response syndrome that arises as a complication of some diseases or infections, and is known to occur as an adverse effect of some antibody drugs and adoptive T-cell therapies. Severe cases have been also been referred to as "cytokine storms”.
  • a priming dose administered as a first administration followed by a full dose of an antibody that binds CD3 and CLEC12A was effective and well tolerated by subjects that were treated for a CLEC12A positive cancer.
  • priming with a lower dose that is escalated to the full dose in one or more steps is instrumental in this.
  • a priming dose is typically given once but can be given twice, or more times in as many administrations.
  • a priming dose can be followed by step- up dosing with gradual increments of the dose until the full dose is reached.
  • a first amount of antibody is preferably a priming dose.
  • Administrations of antibody doses are typically given at regular intervals.
  • the interval between priming and subsequent priming or step-up administrations can vary between 1-7 days, but is typically one day, or three days.
  • the interval between subsequent administrations of step-up doses, if more than one can vary between 3-7 days, but is typically three days.
  • the interval between administrations of a full dose is typically a week.
  • the interval between a first priming administration and a subsequent step-up administration, if any, is typically three days.
  • the interval between the last step-up administration and a subsequent full dose administration, if any, can vary between 3-7 days, but is typically three days.
  • the interval between a first administration and a subsequent administration of a full dose is typically seven days.
  • An administration of a dose is typically calculated per day.
  • a priming dose, a step up dose or a full dose is an amount given at one day.
  • the actual administration may take some time. It is typically but not necessarily an infusion over a period of one or more hours.
  • a priming dose is not given to quickly find a therapeutic dose and to avoid exposure of patients to subtherapeutic doses. Instead it is given to subjects as part of a dosing regimen that is designed to treat the subjects while simultaneously reducing the incidence and/or extent of side effects in the treated population when the full dose is reached.
  • the invention provides a method of treating a subject for a CLEC12A positive cancer the method comprising treating the subject in need thereof with two or more administrations of a bispecific antibody that binds CDS and CLEC12A wherein in a first administration a first amount of the bispecific antibody is administered and wherein in subsequent administrations the amount of bispecific antibody is higher than the amount of bispecific antibody in the first
  • the amount of administered bispecific antibody in the second of said two or more administrations is preferably higher than the amount of bispecific antibody in the first administration, and, in case of more than two administrations, lower than the amount of bispecific antibody in a third of said two or more administrations when the second administration involves a step-up dose. If the second administration is a full dose, the amount of administered bispecific antibody in the second and third administrations is equal. Preferably the amount of administered bispecific antibody in the third of said two or more administrations is higher than the amount of bispecific antibody in the second of said two or more administrations, and, in case where the third administration involves a step-up dose, lower than the amount in a fourth of said two or more administrations.
  • the bispecific antibody is preferably administered at an essentially constant dose in each of the subsequent administrations.
  • the essentially constant dose is also referred to as the full dose.
  • the essentially constant dose in each of the subsequent administrations is preferably at least 15 mg of said bispecific antibody.
  • the essentially constant dose is preferably at least, or equal to, 20, 25, 30, 35, 40, 45, 50, 55 mg, and preferably between 60-600 mg, such as between 60-120; 60-180; 60-200; 60-240; 60-480; 120- 240; 120-480; 240-480; or 240-600 mg, of said bispecific antibody per subsequent administration.
  • the first amount of the bispecific antibody is preferably 9 mg or less, such as between 1-9 mg or between 1-5 mg, preferably 3 mg or less, such as between 1 -3 mg, more preferably about 1 mg, even more preferably about 3 mg, most preferably about 5 mg.
  • the administrations of incremental amounts of antibody are preferably done with an interval of 3 to 4 days between administrations.
  • the interval between the day of administration of the essentially constant amounts of antibody is preferably 6-8 days, preferably 7 days.
  • the first amount of bispecific antibody is preferably a subtherapeutic amount of bispecific antibody.
  • a subtherapeutic amount of bispecific antibody is an amount of antibody that typically does not produce a therapeutic effect when administered in weekly administrations.
  • a first amount of bispecific antibody of between 1-9 mg is given on day 1 of treatment.
  • a first amount of bispecific antibody of 1 mg is given on day 1 of treatment.
  • a first step up dose of 3 mg is given on day 4 of treatment; a second step up dose of 15 mg is given on day 8 of treatment; and a full dose of 25 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a step up dose of 15 mg is given on day 4 of treatment; and a full dose of 25 mg is given on day 8 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a step up dose of 15 mg is given on day 4 of treatment; and a full dose of 25 mg is given on day 8 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 40 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 26 mg is given on day 8 of treatment, a full dose of 40 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 40 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment and a full dose of 40 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 60 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 60 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 60 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment; and a full dose of 60 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 120 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 120 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 120 mg 1 ⁇ 2 given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 120 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 240 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 240 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 240 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment: a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 240 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 400 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 400 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment, a full dose of 400 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment
  • a second step up dose of 25 mg is given on day 8 of treatment
  • a full dose of 400 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment
  • a second step up dose of 25 mg is given on day 8 of treatment
  • a full dose of 600 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 3 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment; a second step up dose of 25 mg is given on day 8 of treatment a full dose of 600 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 10 mg is given on day 4 of treatment
  • a second step up dose of 25 mg is given on day 8 of treatment
  • a full dose of 600 mg is given on day 15 of treatment.
  • a first amount of bispecific antibody of 5 mg is given on day 1 of treatment.
  • a first step up dose of 15 mg is given on day 4 of treatment
  • a second step up dose of 25 mg is given on day 8 of treatment
  • a full dose of 600 mg is given on day 15 of treatment.
  • the dosage regimen can be adapted to include one or more further step-up doses, of for instance 120 mg. It can also be decided to prolong the intervals between the step-up doses and/or the full dose.
  • the invention uses a specific method of administration, preferably with discrete time intervals.
  • the regimen of administration is designed to give an efficacious and safe full dose.
  • a safe dose aims to prevent and minimize the severity and incidence of Cytokine release syndrome (CRS).
  • CRS Cytokine release syndrome
  • a preferred administration regimen comprises administering a priming dose in one or more administrations, one or more step-up doses wherein the dose is increased with increments of the dose until the full dose is reached.
  • the increments are preferably gradual. In one aspect the increments are equal increments until the full dose or maintenance dose is reached. It has, however, been found that starting with a higher priming dose and/or higher first step-up dose leads to less severe side effects, such as for instance cytokine release, at full dose as compared to starting at a low or very low priming dose and/or first step-up dose. In other words, starting with a higher priming dose mitigates cytokine release more effectively than starting with a lower priming dose.
  • Exemplary higher priming doses are for instance 3 or 6 mg.
  • a lower priming dose is for instance 1 mg.
  • the full dose is typically fixed over the treatment period but can be adjusted up or down depending on the individual therapeutic and/or side effect response of the subject.
  • the one or more step-up doses are preferably given in the first one or two weeks of administration.
  • the first priming administration(s) and the step-up administrations are given every 3 days (D1-D4-D8) which is followed by administrations of the full dose every 7 days (on weekly basis).
  • the dose at day 8 (D8) can be equal to the full dose.
  • the full dose is preferably started from day 8-16 depending on the number of step-up administrations.
  • one or more administrations of antibody are accompanied by a pre- and/or concomitant treatment with acetaminophen and an anti-histamine.
  • the anti-histamine is preferably an Hl-antagonist.
  • An Hl- antagonist inhibits the action of histamine at the Hi receptor.
  • the treatment with acetaminophen and an anti-histamine is preferably given on the same day as and preferably prior to the antibody administration.
  • acetaminophen and an anti-histamine is preferably given with each antibody administration.
  • a bispecific antibody that binds CD3 and CLEC12A for use in a method of treatment of CLEC 12A positive cancer in a subject, wherein said treatment comprises two or more administrations of a bispecific antibody that binds CD3 and CLEC12A, wherein in a first administration a first amount of the bispecific antibody is administered and wherein in each of the subsequent administrations the amount of bispecific antibody is higher than the amount of bispecific antibody in the first administration.
  • the invention further provides the use of a bispecific antibody that binds CD3 and CLEC12A for the manufacture of a medicament for the treatment of CLEC12A positive cancer in a subject, wherein in a first administration a first amount of the bispecific antibody is administered and wherein in each of the subsequent administrations the amount of bispecific antibody is higher than the amount of bispecific antibody in the first
  • a method of pinging CLEC12A positive hemopoietic cells from a subject and repopulating the hemopoietic system of said subject with normal CLEC12A positive cells comprising administering to the subject in need thereof a bispecific antibody that binds CD3 and CLEC12A at intervals thereby stimulating hemopoietic stem cells of said subject to repopulate said hemopoietic system with newly formed hemopoietic stem cell derived cells, including CLEC12A positive cells.
  • the subject is preferably a subject with AML, myeloproliferative neoplasm blast phase (MPN-BP), myelofibrosis (MF) or myelodysplastic syndrome (MDS).
  • the subject has myeloproliferative neoplasm blast phase (MPN-BP), myelofibrosis (MF) or myelodysplastic syndrome (MDS).
  • MPN-BP myeloproliferative neoplasm blast phase
  • MF myelofibrosis
  • MDS myelodysplastic syndrome
  • Normal hemopoietic stem cells were found to be predominantly CLEC12A negative.
  • CLEC12A expression is restricted to the granulocyte-macrophage progenitor (GMP) cells and a fraction of the common myeloid progenitor (CMP) and megakaryocyte-erythroid progenitor (MEP) cells.
  • GMP granulocyte-macrophage progenitor
  • CMP common myeloid progenitor
  • MEP megakaryocyte-erythroid progenitor
  • CMP and MEP that are not purged can still give rise to GMP and BFU-E (erythrocytes) and CFU- Mk (megakaryocytes).
  • Treatment with a CD3xCLECl2A bispecific antibody as described herein can remove malignant cells, while leaving the potential of the hemopoietic stem and progenitor cells untouched. These are stimulated upon treatment with an antibody as described herein to repopulate the hemopoietic system with new, healthy cells, including CLEC12A positive cells. The stimulated cells also produce new differentiated CLEC12A negative cells.
  • Repopulation time is at least shorter than with other CDS engager approaches such as CD19 and CD123.
  • CFU-Mk and BFU-E progenitors are spared in a treatment of the invention the platelet producing capacity and the erythrocyte producing capacity of the hemopoietic system are also spared when compared to other CD3 engager formats.
  • a method for stimulating granulocyte and monocyte development in a subject comprising administering a bispecific antibody that binds CD3 and CLEC12A at intervals to the subject in need thereof thereby inducing the development of granulocytes and monocytes from CLEC12A negative hemopoietic stem cells or progenitor cells.
  • the administration at intervals preferably follows the interval as described herein above.
  • the dosing schedule is preferably as described herein.
  • the subject is preferably a subject with AML, myeloproliferative neoplasm blast phase (MPN-BP), myelofibrosis (MF) or myelodysplastic syndrome (MDS).
  • MPN-BP myeloproliferative neoplasm blast phase
  • MF myelofibrosis
  • MDS myelodysplastic syndrome
  • Also provided is a method of preparing an autologous bone marrow cell graft for a subject that is being treated for a CLEC12A positive cancer comprising incubating a bone marrow cell preparation of said subject with a bispecific antibody that binds CD3 and CLEC12A under conditions suitable for killing of CLEC12A positive cells and subsequently collecting bone marrow cells from said incubation.
  • a method of treating a CLEC12A positive cancer in a subject the method comprising incubating a bone marrow cell preparation of said subject with a bispecific antibody that binds CD3 and
  • CLEC12A under conditions suitable for killing of CLEC12A positive cells, treating the subject with an hemopoietic system ablative therapy and providing said subject with a bone marrow cell graft comprising said incubated bone marrow cells.
  • T-cells in the autologous graft derived from the bone marrow or the blood in the graft are retargeted to eliminate CLEC12A positive cells from the autologous graft.
  • a CDS and CLEC12A bispecific antibody as disclosed herein is effective even at low effector target cell ratio’s, including at a range of 1:3-1:97, and at levels reflected in Fig 6 and Table II.
  • Conditions suitable for killing of CLEC12A positive cells as mentioned herein entail the presence of T cells such as autologous T cells or preactivated T cells. Such cells are normally present in a bone marrow preparation as a result of the presence of a significant amount of blood that is typically also collected with many present day bone marrow harvest methods. Ablative therapy is a treatment that results in the destruction of cells as a result of the therapy. Many chemotherapies and other anti-tumor therapies also kill normal cells. One of the more sensitive organs is the bone marrow.
  • a hemopoietic system ablative therapy in the context of the present invention is a therapy (or treatment) that has the additional or side-effect that a major part of the“normal” or“healthy” hemopoietic system is killed by the therapy or treatment.
  • Bone marrow is often collected prior to treating the patient with the hemopoietic system ablative therapy. This bone marrow is transplanted back into the subject after completion of the therapy to stimulate the repopulation of the hemopoietic system of the subject. To prevent that aberrant cells are also transplanted, such bone marrow preparations are typically purged for aberrant cells. Sorting CD34 positive cells is one method when aberrant cells are not CD34 positive.
  • a method of purging of the present invention efficiently removes aberrant cells from the bone marrow preparation allowing quicker repopulation.
  • a method of providing a subject with a hemopoietic stem cell sparing cancer treatment comprises administering a bispecific antibody that binds CD3 and CLEC12A to the subject in need thereof.
  • the subject has a CLEC12A positive cancer.
  • a bispecific antibody as disclosed herein is preferably administered as an intravenous infusion. Infusions typically take about 2-4 hours but longer and shorter administrations are also possible. Infusions do not normally extend over a period of more than 12 hours.
  • the increments in the amount of antibody until the essentially constant amount per administration is reached is also referred to as a dose escalation regimen within a single patient, or intra -patient dose escalation.
  • CLEC12A refers to C type lectin domain family 12 member A.
  • CLEC12A is also referred to as C-Type Lectin Protein CLL-1; MI CL; Dendritic Cell-Associated Lectin 2; C-Type Lectin Superfamily; Myeloid Inhibitory C-Type Lectin-Like Receptor; C-Type Lectin-Like Molecule-1; DCAL2; CLLl; C- Type Lectin-Like Molecule 1; DCAL-2; Killer cell lectin like receptor subfamily L, member 1 (KLRLl); CD371(cluster of differentiation 371) (Bakker, van den
  • CLEC12A is an antigen that is expressed on leukemic blast cells and on leukemic stem cells in acute myeloid leukemia (AML), including the CD34 negative or CD34 low expressing leukemic stem cells (side population) (Bakker, van den Oudenrijn et al. 2004; van Rhenen, van Dongen et al. 2007; Moshaver, van Rhenen et al, 2008), as well as in myelodysplastic syndromes (MDS) (Bakker, van den Oudenrijn et al. 2004, and Toft-Peterson, Nederby et al, 2016).
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndromes
  • CLEC12A is otherwise thought to be restricted to cells of the hemopoietic lineage, particularly to myeloid lineage in peripheral blood and bone marrow, i.e., granulocytes, monocytes and dendritic cell precursors. More importantly,
  • CLEC12A is absent on normal hemopoietic stem cells. Where reference is made to CLEC12A herein, the reference is to human CLEC12A (SEQ ID NO: 1), unless specifically stated otherwise.
  • CLAG 12 A means all variants (such as splice and mutation) that are referenced herein and isoforms thereof that retain the myeloid expression profile (both at surface expression level and mRNA level) including as described in Bakker, van den Oudenrijn et al. 2004 and Marshall, Willment et al. 2004. While accession numbers are primarily provided as a further method of identification, the actual sequence of the protein may vary, for instance because of a mutation in the encoding gene such as those occurring in some cancers or the like.
  • CDS cluster of differentiation 3
  • CD3y chain SwissFrot F09693
  • CD38 chain SwissProt
  • CD3c is known under various aliases some of which are:
  • CD3e Molecule Epsilon (CD3-TCR Complex)
  • CD3e Antigen Epsilon
  • T3 Complex Polypeptide (TiT3 Complex)”; T-Cell Surface Antigen T3/Leu-4 Epsilon Chain; T3E; T-Cell Antigen Receptor Complex, Epsilon Subunit Of T3; CD3e Antigen; CD3- Epsilon 3; IMD18; TCRE.
  • Ids for CD3E Gene are HGNC: 1674; Entrez Gene: 916; Ensembl: ENSG00000198851; OMIM: 186830 and UniProtKB: P07766.
  • TCR T-cell receptor
  • the TCR, x-chain, and CD3 molecules together comprise the TCR complex.
  • CD 3 is expressed on T cells. Where reference is made to CD3 herein, the reference is to human CDS (SEQ ID NOs: 2-5), unless specifically stated otherwise.
  • antibody as used herein means a proteinaceous molecule belonging to the immunoglobulin class of proteins, containing one or more domains that bind an epitope on an antigen, where such domains are or derived from or share sequence homology with the variable region of an antibody.
  • Antibodies are typically made of basic structural units - each with two heavy chains and two light chains.
  • Antibodies for therapeutic use are preferably as close to natural antibodies of the subject to be treated as possible (for instance human antibodies for human subjects).
  • An antibody according to the present invention is not limited to any particular format or method of producing it.
  • A“bispecific antibody” is an antibody as described herein wherein one domain of the antibody binds to a first antigen whereas a second domain of the antibody binds to a second antigen, wherein said first and second antigens are not identical.
  • the term“bispecific antibody” also encompasses antibodies wherein one heavy chain variable region/light chain variable region (VH/VL) combination binds a first epitope on an antigen and a second VH/VL combination that binds a second epitope.
  • VH/VL heavy chain variable region/light chain variable region
  • the term further includes antibodies wherein VH is capable of specifically recognizing a first antigen and the VL, paired with the VH in an immunoglobulin variable region, is capable of specifically recognizing a second antigen.
  • VH/VL pair will bind either antigen 1 or antigen 2.
  • antigen 1 or antigen 2 Such so called“two- in-one antibodies”, described in for instance WO 2008/027236, WO 2010/108127 and Schaefer, Haber et al, 2011.
  • a bispecific antibody according to the present invention is not limited to any particular bispecific format or method of producing it.
  • the term‘common light chain’ as used herein refers to the two light chains (or the VL part thereof) in the bispecific antibody.
  • the two light chains (or the VL part thereof) may be identical or have some amino acid sequence differences while the binding specificity of the full length antibody is not affected.
  • “Common” also refers to functional equivalents of the light chain of which the amino acid sequence is not identical. Many variants of said light chain exist wherein mutations
  • the light chain of the present invention can also be a light chain as specified herein above, having from 0 to 10, preferably from 0 to 5 amino acid insertions, deletions, substitutions, additions or a combination thereof. It is for instance within the scope of the definition of common light chains as used herein, to prepare or find light chains that are not identical but still functionally equivalent, e.g., by introducing and testing conservative amino acid changes, changes of amino acids in regions that do not or only partly contribute to binding specificity when paired with the heavy chain, and the like.
  • full length IgG or‘full length antibody * according to the invention 1 ⁇ 2 defined as comprising an essentially complete IgG, which however does not necessarily have all functions of an intact IgG.
  • a full length IgG contains two heavy and two light chains. Each chain contains constant (C) and variable (V) regions, which can be broken down into domains designated CHI, CH2, CH3, VH, and CL, VL.
  • An IgG antibody binds to antigen via the variable region domains contained in the Fab portion, and after binding can interact with molecules and cells of the immune system through the constant domains, mostly through the Fc portion.
  • Full length antibodies according to the invention encompass IgG molecules wherein mutations may be present that provide desired characteristics.
  • Full length IgG should not have deletions of substantial portions of any of the regions.
  • IgG molecules wherein one or several amino acid residues are deleted, without essentially altering the binding characteristics of the resulting IgG molecule are embraced within the term "full length IgG".
  • such IgG molecules can have a deletion of between 1 and 10 amino acid residues, preferably in non-CDR regions, wherein the deleted amino acids are not essential for the binding specificity of the IgG.
  • Percent (%) identity as referred to amino acid sequences herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a selected sequence, after aligning the sequences for optimal comparison purposes. In order to optimize the alignment between the two sequences gaps may be introduced in any of the two sequences that are compared. Such alignment can be carried out over the full length of the sequences being compared. Alternatively, the alignment may be carried out over a shorter length, for example over about 20, about 50, about 100 or more nucleic acids/based or amino acids. The sequence identity is the percentage of identical matches between the two sequences over the reported aligned region.
  • a comparison of sequences and determination of percentage of sequence identity between two sequences can be accomplished using a mathematical algorithm.
  • the skilled person will be aware of the fact that several different computer programs are available to align two sequences and determine the identity between two sequences (Kruskal, J. B., 1983).
  • the percent sequence identity between two amino acid sequences may be determined using the Needleman and Wunsch algorithm for the alignment of two sequences. (Needleman and Wunsch,
  • the Needleman- Wunsch algorithm has been implemented in the computer program NEEDLE.
  • the NEEDLE program from the EMBOSS package may be used (version 2.8.0 or higher, Rice, Longden et al, 2000; http://embos8.bioinformatics.nl/).
  • EBLOSUM62 is used for the substitution matrix.
  • the optional parameters used are a gap-open penalty of 10 and a gap extension penalty of 0.5.
  • the percentage of sequence identity between a query sequence and a sequence of the invention is calculated as follows: Number of corresponding positions in the alignment showing an identical amino acid or identical nucleotide in both sequences divided by the total length of the alignment after subtraction of the total number of gaps in the alignment.
  • antibodies according to the present invention that“specifically recognize” an antigen, for example, CLEC12A or CD3, may recognize other compounds as well, if such other compounds contain the same kind of epitope.
  • the terms“specifically recognizes’’ with respect to an antigen and antibody interaction does not exclude binding of the antibodies to other compounds that contain the same kind of epitope.
  • epitopes refers to a site on an antigen to which an immunoglobulin or antibody specifically binds.
  • Epitopes can be formed both from contiguous amino acids or noncontiguous amino adds juxtaposed by tertiary folding of a protein (so-called linear and conformational epitopes). Epitopes formed from contiguous, linear amino adds are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding, conformation are typically lost on treatment with denaturing solvents.
  • An epitope may typically include 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes are known to persons of ordinary skill in the art and include techniques in the art for example, x-ray crystallography, HDX-MS and 2-dimensional nuclear magnetic resonance, pepscan, and alanine scan depending on the nature of the epitope (see, e.g., Morris G.E., 1996).
  • tumor cells includes tumor cells, more specifically tumor cells of hematological origin including also pre-leukemic cells such as cells that cause myelodysplastic syndromes (MDS) and leukemic cells such as acute myeloid leukemia (AML) tumor cells or chronic myelogenous leukemia (CML) cells.
  • MDS myelodysplastic syndromes
  • leukemic cells such as acute myeloid leukemia (AML) tumor cells or chronic myelogenous leukemia (CML) cells.
  • AML acute myeloid leukemia
  • CML chronic myelogenous leukemia
  • Immune effector cell refers to a cell within the natural repertoire of cells in the mammalian immune system which can be activated to affect the viability of a target cell.
  • Immune effector cells include cells of the lymphoid lineage such as natural killer (NK) cells, T cells including cytotoxic T cells, or B cells, and including cells of the myeloid lineage, such as monocytes or macrophages, dendritic cells and neutrophilic granulocytes.
  • NK natural killer
  • T cells including cytotoxic T cells, or B cells
  • myeloid lineage such as monocytes or macrophages, dendritic cells and neutrophilic granulocytes.
  • said effector cell is preferably an NK cell, a T cell, a B cell, a monocyte, a
  • effector cells to aberrant cells means that immune effector cells are brought in proximity to the aberrant target cells such that the effector cells can directly kill, or indirectly initiate the killing of the aberrant cells.
  • the terms "subject” and “patient” are used interchangeably and refer to a mammal such as a human, mouse, rat, hamster, guinea pig, rabbit, cat, dog, monkey, cow, horse, pig and the like (e.g., a patient, such as a human patient, having cancer).
  • treat refers to any type of intervention or process performed on, or administering an active agent or combination of active agents to the subject with the objective of reversing, alleviating, ameliorating, inhibiting, or slowing down or preventing the
  • a beneficial effect can take the form of an improvement over baseline, including an improvement over a measurement or observation made prior to initiation of therapy according to the method.
  • a beneficial effect can take the form of slowing, stabilizing, stopping or reversing the progression of a cancer in a subject at any clinical stage, as evidenced by a decrease or elimination of a clinical or diagnostic symptom of the disease, or of a marker of cancer.
  • Effective treatment may, for example, decrease in tumor size, decrease the presence of circulating tumor cells, reduce or prevent metastases of a tumor, slow or arrest tumor growth and/or prevent or delay tumor recurrence or relapse.
  • therapeutic amount refers to an amount of an agent or combination of agents that provides the desired biological, therapeutic, and/or prophylactic result. That result can be reduction, amelioration, palliation, lessening, delaying, and/or alleviation of one or more of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • a therapeutic amount 1 ⁇ 2 an amount sufficient to delay tumor development.
  • a therapeutic amount is an amount sufficient to prevent or delay tumor recurrence.
  • a therapeutic amount can be administered in one or more administrations.
  • the therapeutic amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and may stop cancer cell infiltration into peripheral organs; (iv) inhibit tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor, and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
  • an“therapeutic amount” is the amount of a CLEC12A/CD3 bispecific antibody that effects a decrease in a cancer (for example a decrease in the number of cancer cells) or slowing of progression of a cancer, such as acute myeloid leukemia, myelodysplastic syndrome or chronic myelogenous leukemia.
  • bispecific antibodies for use in the methods provided herein include bispecific antibodies that comprise one heavy chain variable region/light chain variable region (VH/VL) combination that binds CLEC12A, and a second VH/VL combination that binds CD3.
  • VH/VL heavy chain variable region/light chain variable region
  • CLEC12A heavy chain variable (VH) regions for use in a
  • CLEC12A/CD3 bispecific antibody include those which bind to CLEC12A.
  • the CLEC12A VH region of a CLEC12A/CD3 bispecific antibody binds to CLEC12A expressed on tumor cells.
  • Exemplary CLEC12A VH regions for use in the CLEC12A/CD3 bispecific antibody are disclosed, for example, in W02017/010874, WO2014/051433, and W02005/000894 (each of which is incorporated herein by reference).
  • the binding affinity of the CLEC12A/CD3 bispecific antibody for CLEC12A on tumor cells is at least 2 times, 4 times, 6 times, 10 times, 20 times, 30 times, 40 times or 50 times higher than the affinity of binding to CDS.
  • the CLEC12A binding affinity of the CLEC12A/CD3 bispecific antibody for CLEC12A is between about lxlOe-6 M and lxlOe-10 M, between about lxlOe-7 M and lxlOe-10 M, or between about lxlOe-8 and lxlOe-10.
  • CLEC12A binding affinity of the CLEC12A/CD3 bispecific antibody for CLEC12A is at least Ixl0-e8 M, preferably at least Ixl0-e9 M.
  • the binding affinity of the CLEC12A/CD3 bispecific antibody for CLEC1.2A is between about lxlOe-8 M and lxlOe-9 M including, for example, about 2xl0e-9 M, about 3xl0e-9 M, about 4xl0e-9 M, about 5xl0e-9 M, about 6xl0e-9 M, about 7xl0e-9 M, 8xl0e-9 M or about 9xl0e-9 M, and the binding affinity of the CLEC12A/CD3 bispecific antibody for CDS is at least 30 times lower, at least 40 times lower, or at least 50 times lower.
  • the CDS VH region of the CLEC12A/CD3 bispecific antibody binds to cell surface expressed CD3/TCR on human T cell lines with an affinity (KD) that is significantly less than the affinity of murine anti-CDS antibody, mOKT3.
  • the CDS VH region of the CLEC12A/CD3 bispecific antibody binds to CD3 with a binding affinity of at least Ixl0-e6 M.
  • the CD3 binding affinity of the bispecific antibody is between about lxlOe-6 M and lxlOe-10 M.
  • the CD3 binding affinity of the CD3 VH region of the bispecific antibody is between about lxl Oe- 7 M - lxlOe-8 M.
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the heavy chain variable region comprises:
  • a heavy chain CDRl comprising the amino acid sequence SGYTFTGY (SEQ ID NO: 9), a heavy chain CDR2 comprising the amino arid sequence IINPSGGS (SEQ ID NO: 10-), and a heavy chain CDR3 comprising the amino acid sequence GTTGDWFDY (SEQ ID NO: 11);
  • a heavy chain CDRl comprising the amino acid sequence SGYTFTSY (SEQ ID NO: 13), a heavy chain CDR2 comprising the amino acid sequence IINPSGGS (SEQ ID NO: 14), and a heavy chain CDR3 comprising the amino acid sequence GNYGDEFDY (SEQ ID NO: 15);
  • a heavy chain CDRl comprises the amino arid sequence SGYTFTGY (SEQ ID NO: 17), a heavy chain CDR2 comprising the ammo acid sequence WINPNSGG (SEQ ID NO: 18), and a heavy chain CDR3 comprising the amino acid sequence DGYFADAFDY (SEQ ID NO: 19).
  • said heavy chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one ammo acid from the recited CDR sequences.
  • the heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CLEC12A, wherein said heavy chain variable region comprises the HCDRl, HCDR2 and HCDR3 of the VH region set forth in SEQ ID NOs: 12, 16 or 20.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CLEC12A, wherein said heavy chain variable region comprises an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino arid sequence set forth in SEQ ID NOs: 12, 16 or 20.
  • the heavy chain variable region of the bispecific antibody that binds human CLEC12A can have 0-10, preferably 0-5 amino acid insertions, deletions, substitutions, additions in the sequence of the heavy chain variable region outside of the three CDR sequences, or a combination thereof.
  • the heavy chain variable region comprises from 0 to 9, from 0 to 8, from 0 to 7, from 0 to 6, from 0 to 5, from 0 to 4, preferably from 0 to 3, preferably from 0 to 2, preferably from 0 to 1 and preferably 0 amino acid insertions, deletions, substitutions, additions with respect to the indicated ammo acid sequence, or a combination thereof.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CLEC12A, wherein said heavy chain variable region comprises an amino acid sequence selected from SEQ ID NOs: 12,
  • VH heavy chain variable
  • CLEC12A/CD3 bispecific antibody include those which can bind a CD3y chain, a CD38 chain, a CD3e chain or a combination of CD36/CD3e or CD3y/CD3e.
  • the CD3 VH region of the bispecific antibody binds the CD3e chain.
  • the CD3 VH region binds to human CD3.
  • the CD3 VH region binds to the human CD3e chain.
  • CD3 binding regions for use in the CLEC12A/CD3 bispecific antibody are disclosed in W02017/010874, WO2014/061433 and WO2005/118635 (each of which is incorporated herein by reference).
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds CD3, wherein said heavy chain variable region comprises:
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYNGRKQ (SEQ ID NO: 22), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP(SKQ ID NO: 23);
  • a heavy chain CDRl comprising the ammo acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYSGSKKN(SEQ ID NO: 30), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWYHGRKQ (SEQ ID NO: 32), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYHARKQ (SEQ ID NO: 34), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWYNARKQ (SEQ ID NO: 36), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYNTRKQ (SEQ ID NO: 45), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the ammo acid sequence IWYDGKNT (SEQ ID NO: 47), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IYYDGSRT (SEQ ID NO: 49), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23); or
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWHDGRKT (SEQ ID NO: 51), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23).
  • the CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21) is defined according to IMGT.
  • the CDRl may comprise the amino add sequence SYGMH (SEQ ID NO: 60) as defined according to Kabat.
  • variable amounts of 1, 2 or 3 amino add residues from the recited CDR sequences are allowed while retaining the same kind of binding activity (in kind, not necessarily in amount).
  • said heavy chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one ammo add from the recited CDR sequences.
  • the heavy chain CDR 1, 2 and 3 sequences are identical to the redted CDR sequences.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CDS, wherein said heavy chain variable region comprises the HCDR1, HCDR2 and HCDR3 of the VH region set forth in SEQ ID NOs: 24-29, 31, 33, 35, 37-44, 46, 48, 50 and 52.
  • the CLEC12AZCD3 bispecific antibody comprises a heavy chain variable region that binds human CD3, wherein said heavy chain variable region comprises at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino add sequence of one of the VH region sequences set forth in SEQ ID NO: 24-29, 31, 33, 35, 37-44, 46, 48, 50 and 52.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CD3, wherein said heavy chain variable region comprises at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino acid sequence of one of the sequences VH region set forth in SEQ ID NO: 37-44.
  • the heavy chain variable region of the bispecific antibody that binds human CD3 can have from 0 to 10, preferably from 0 to d amino acid insertions, deletions, substitutions, additions in the sequence of the heavy chain variable region outside of the three CDR sequences, or a combination thereof.
  • the heavy chain variable region comprises from 0 to 9, from 0 to 8, from 0 to 7, from 0 to 6, from 0 to 5, from 0 to 4, preferably from 0 to 3, preferably from 0 to 2, preferably from 0 to 1 and preferably 0 amino acid insertions, deletions, substitutions, additions with respect to the indicated amino acid sequence, or a combination thereof.
  • Additional variants of the disclosed amino acid sequences which retain CLEC12A or CD3 binding can be obtained, for example, from phage display libraries which contain the rearranged human IGKV1-39/IGKJ1 VL region (de Kruif, Kramer et al. 2010), and a collection of VH regions incorporating ammo acid substitutions into the amino acid sequence of a CLEC12A or CD3 VH region disclosed herein, as previously described (e.g., US 2016/0368988).
  • Phages encoding Fab regions which bind CLEC12A or CD3 may be selected and analyzed by flow cytometry, and sequenced to identify variants with amino acid substitutions, insertions, deletions or additions which retain antigen binding.
  • the CD3 VH region may be substituted at position A50 and be modified by an S, Y, M or a Q; D59 may be substituted by L, I, V, F, R, A, N, H, S, T, Y or E, preferably by an Y or an E; A61 may be substituted by N, I,
  • H, Q, L, R, Y, E, S, T, D, K, V; and F105 may be substituted by an Y or an M.
  • the CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CD3, wherein said heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 24-29, 31, 33, 35, 37-44, 46, 48, 50 or 52.
  • CLEC12A/CD3 bispecific antibody comprises a heavy chain variable region that binds human CDS, wherein said heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 37-44.
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the first VH region comprises an amino acid sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino acid sequence of the VH region set forth in SEQ ID NO: 12, 16 and 20; and a second heavy chain variable region that binds human CD3 comprising, wherein the second VH region comprises an amino acid sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino arid sequence of one of the VH region sequences set forth in SEQ ID NO: 37-44.
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, and a second heavy chain variable region that binds human CD
  • the first heavy chain variable region comprises:
  • SGYTFTGY (SEQ ID NO: 9), a heavy chain CDR2 comprising the amino acid sequence IINPSGGS (SEQ ID NO: 10), and a heavy chain CDR3 comprising the amino acid sequence GTTGDWFDY (SEQ ID NO: 11);
  • SGYTFTSY (SEQ ID NO: 13), a heavy chain CDR2 comprising the amino acid sequence IINPSGGS (SEQ ID NO: 14), and a heavy chain CDR3 comprising the amino acid sequence GNYGDEFDY (SEQ ID NO: 15); or
  • a heavy chain CDRl comprises the amino acid sequence
  • SGYTFTGY (SEQ ID NO: 17), a heavy chain CDR2 comprising the amino acid sequence WINPNSGG (SEQ ID NO: 18), and a heavy chain CDR3 comprising the amino acid sequence DGYFADAFDY (SEQ ID NO: 19);
  • the second heavy chain variable region comprises,
  • GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYNGRKQ (SEQ ID NO: 22), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP(SEQ ID NO: 23);
  • GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYSGSKKN (SEQ ID NO: 30), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYHGRKQ (SEQ ID NO: 32), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDR1 comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino acid sequence IWYHARKQ (SEQ ID NO: 34), and a heavy chain CDR3 comprising the ammo acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWYNARKQ (SEQ ID NO: 36), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDR1 comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWYNTRKQ (SEQ ID NO: 46), and a heavy chain CDR3 comprising the ammo acid sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDR1 comprising the ammo add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWYDGKNT (SEQ ID NO: 47), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23);
  • a heavy chain CDRl comprising the amino add sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IYYDGSRT (SEQ ID NO: 49), and a heavy chain CDR3 comprising the ammo acid sequence GTGYNWFDP (SEQ ID NO: 23); or
  • a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 sequence comprising the amino add sequence IWHDGRKT (SEQ ID NO: 51), and a heavy chain CDR3 comprising the amino add sequence GTGYNWFDP (SEQ ID NO: 23).
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the first VH region comprises a heavy chain CDRl comprising the amino add sequence SGYTFTGY (SEQ ID NO: 9), a heavy chain CDR2 comprising the amino add sequence IINPSGGS (SEQ ID NO: 10), and a heavy chain CDR3 comprising the amino acid sequence GTTGDWFDY (SEQ ID NO: 11); and a second heavy chain variable region, wherein the second VH region comprises a heavy chain CDRl comprising the amino acid sequence GFTFSSYG (SEQ ID NO: 21), a heavy chain CDR2 comprising the amino acid sequence IWYNARKQ (SEQ ID NO: 36), and a heavy chain CDR3 comprising the amino acid sequence GTGYNWFDP (SEQ ID NO: 23).
  • the first VH region comprises a heavy chain CDRl comprising the amino add sequence SGYTFTGY (SEQ ID NO: 9), a heavy
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the amino acid sequence of the first VH region is selected from SEQ ID NOs: 12, 16 and 20; and a second heavy chain variable region that binds human CD3, wherein the amino acid sequence of the second VH region is selected from SEQ ID NOs: 24-29, 31, 33, 35, 37-44, 46, 48, 50 and 52.
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the amino acid sequence of the first VH region 1 ⁇ 2 SEQ ID NO: 12; and a second heavy chain variable region that binds human CD3, wherein the amino acid sequence of the second VH region is selected from SEQ ID NOs: 37-44.
  • the CLEC12A/CD3 bispecific antibody comprises a first heavy chain variable region that binds human CLEC12A, wherein the amino acid sequence of the first VH region is set forth in SEQ ID NO: 12; and a second heavy chain variable region that binds human, wherein the amino acid sequence of the second VH region is set forth in SEQ ID NO: 37.
  • the light chain variable regions of the VH/VL CLEC12A binding region and the VH/VL binding region of the CD3 binding region of the CLEC12A/CD3 bispecific antibody may be the same as the VL region of parental CLEC12A monospecific antibody and/or the VL region of parental CD3 monospecific antibody, or alternative VL regions may be used for one or both VH/VL region combinations as long as the bispecific antibody retains binding to both the CLEC12A and CDS antigens.
  • the VL region of the VH/VL CLEG 12 A binding region of the CLEC12AZCD3 bispecific antibody is similar to the VL region of the VH/VL CD3 binding region.
  • VL regions in the first and second VH/VL region combinations are identical.
  • the light chain variable region of one or both VH/VL binding regions of the CLEC12A/CD3 bispecific antibody comprises a common light chain.
  • the common light chain variable region of one or both VH/VL binding regions comprises a germline 012 variable region V- segment.
  • the light chain variable region of one or both VH/VL binding regions comprises the kappa light chain V-segment IgVxl-39*01. IgVal-39 is short for Immunoglobulin Variable Kappa 1-39 Gene.
  • the gene is also known as Immunoglobulin Kappa Variable 1-39; IGKV139; IGKV1-39; 012a or 012.
  • External Ids for the gene are HGNC: 5740; Entrez Gene: 28930; Ensembl: ENSG00000242371.
  • the amino add sequence for the V-region is provided in SEQ ID NO: 56.
  • the V-region can be combined with one of five J -regions.
  • Preferred J- regions are jkl and jk5, and the joined sequences are indicated as IGKVl-39/jkl and IGKVl-39/jko; alternative names are IgVKl-39*01/IGJn:l*01 or IgVxl- 39*01/IGJK5*01 (nomenclature according to the IMGT database worldwide web at imgt.org).
  • the light chain variable region of one or both VH/VL binding regions comprises the kappa light chain IgVKl-39*01/IGJKl*01 or JgVis 1-39*01/IGJK1*06 (SEQ ID NO: 57 and SEQ ID NO: 58, respectively).
  • the light chain variable region of one or both VH/VL binding regions of the CLEC12A/CD3 bispecific antibody comprises an LCDR1 comprising the amino acid sequence QSISSY (SEQ ID NO: 53), an LCDR2 comprising the amino acid sequence AAS, and an LCDR3 comprising the amino acid sequence QQSYSTP (SEQ ID NO: 55) (i.eflower the CDRs of IGKVl-39 according to IMGT).
  • the light chain variable region of one or both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprises an LCDRl comprising the amino acid sequence QSISSY (SEQ ID NO: 53), an LCDR2 comprising the amino acid sequence AASLQS (SEQ ID NO: 54), and an LCDR3 comprising the amino acid sequence QQSYSTP (SEQ ID NO: 56).
  • GLEC12A/CD3 bispecific antibody comprise a light chain variable region comprising an amino add sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino acid sequence of set forth in
  • one or both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprise a light chain variable region comprising an amino acid sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical or 100% identical to the amino add sequence of set forth in SEQ ID NO: 68.
  • variable light chain of one or both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody can have from 0 to 10, preferably from 0 to 5 amino acid insertions, deletions, substitutions, additions or a combination thereof with respect to SEQ ID NO: 57 or SEQ ID NO: 58.
  • the light chain variable region of one or both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprises from 0 to 9, from 0 to 8, from 0 to 7, from 0 to 6, from 0 to 5, from 0 to 4, preferably from 0 to 3, preferably from 0 to 2, preferably from 0 to 1 and preferably 0 amino acid insertions, deletions, substitutions, additions with respect to the indicated amino acid sequence, or a combination thereof.
  • the light chain variable region of one or both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprises the amino acid sequence of SEQ ID NO: 57 or SEQ ID NO: 58 .
  • both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprise identical
  • VL regions In one embodiment, the VL of both VH/VL binding regions of the CLEC12A/CD3 bispedfic antibody comprises the amino acid sequence set forth in SEQ ID NO: 57. In one embodiment, the VL of both VH/VL binding regions of the CLEC12A/CD3 bispecific antibody comprises the amino acid sequence set forth in SEQ ID NO: 58.
  • CLEC12A/CD3 bispecific antibodies for use in the methods disclosed herein can be provided in a number of formats. Many different formats of bispecific antibodies are known in the art, and have been reviewed by Kontermann,
  • bispecific antibody formats that are not classical antibodies with two VH/VL combinations, have at least a variable domain comprising a heavy chain variable region and a light chain variable region.
  • This variable domain may be linked to a single chain Fv-fragment, monobody, a VH and a Fab-fragment that provides the second binding activity.
  • the CLEC12A/CD3 bispecific antibodies used in the methods provided herein are generally of the human IgG subclass (e.g., for instance IgGl, IgG2, IgG3, IgG4). In certain embodiments, the antibodies are of the human IgGl subclass. Full length IgG antibodies are preferred because of their favorable half-life and for reasons of low immunogenicity . Accordingly, in certain
  • the CLEC12AZCD3 bispecific antibody is a full length IgG molecule. In an embodiment, the CLEC12AZCD3 bispecific antibody is a full length IgGl molecule.
  • the CLEC12A/CD3 bispecific antibody comprises a fragment crystallizable (Fc).
  • the Fc of the CLEC12A/CD3 bispecific antibody is preferably comprised of a human constant region.
  • a constant region or Fc of the CLEC12A/CD3 bispecific antibody may contain one or more, preferably not more than 10, preferably not more than 5 amino-acid differences with a constant region of a naturally occurring human antibody.
  • each Fab-arm of the bispecific antibodies may further include an Fc- region comprising modifications promoting the formation of the bispecific antibody, modifications affecting Fc-mediated effector functions, and/or other features described herein.
  • a CLEC12A/CD3 bispecific full length IgG antibody has a mutated lower hinge and/or CH2 domains such that interaction of said bispecific IgG antibody with Fc gamma (Fey) receptors is reduced.
  • Fc gamma Fc gamma
  • the term“such that interaction of said bispecific IgG antibody with Fc gamma receptors is reduced” means that the interaction of the CLEC12A/CD3 bispecific antibody with Fc gamma receptors, if such Fc gamma receptors are present in the vicinity of the antibody, is reduced.
  • the interaction of the CLEC12A/CD3 bispecific antibody with the Fc receptor is essentially abolished.
  • Bispecific antibodies with reduced Fey receptor binding have been previously described (US 2014/0120096, incorporated herein by reference).
  • the CLEC12A/CD3 bispecific antibody comprises a mutated lower hinge and/or CH2 domain with at least one substitution at amino acids positions 235 and/or 236 (EU numbering). Preferably, both amino acids positions 235 and 236 are substituted. As described in US 2014/0120096, substitutions at these sites are capable of essentially preventing the interaction between an antibody and the Fc receptor present on tumor cells or effector cells. Accordingly, in certain embodiments, the CLEC12A/CD3 bispecific antibody comprises a mutated CH2 and/or lower hinge domains comprising an L235G and/or G236E substitution. Preferably, both L235G and G236R are substituted.
  • Bispedfic antibodies are typically produced by cells that express nucleic acid(s) encoding the antibody. Accordingly, in some embodiments, the bispedfic CLEC12A/CD3 antibodies disdosed herein are produced by providing a cell comprising one or more nucleic adds that encode the heavy and light chain variable regions and constant regions of the bispedfic CLEC12A/CD3 antibody.
  • the cell is preferably an animal cell, more preferably a mammal cell, more preferably a primate cell, most preferably a human cell.
  • a suitable cell is any cell capable of comprising and preferably of producing the CLEC12A/CD3 bispedfic antibody.
  • Suitable cells for antibody production include a hybridoma cell, a Chinese hamster ovary (CHO) cell, an NSO cell or a PER-C6 cell.
  • CHO Chinese hamster ovary
  • NSO Chinese hamster ovary
  • PER-C6 PER-C6 cells
  • Various institutions and companies have developed cell lines for the large scale production of antibodies, for instance for clinical use.
  • Non-limiting examples of such cell lines are CHO cells, NSO cells or PER.C6 cells.
  • said cell is a human cell.
  • a preferred example of such a cell line is the PER.C6 cell line or equivalent thereof.
  • said cell is a CHO cell or a variant thereof.
  • GS Glutamine synthetase
  • the cell is a CHO cell.
  • the cell expresses the different light and heavy chains that make up the CLEC12A/CD3 bispecific antibody. In certain embodiments, the cell expresses the different light and heavy chains that make up the CLEC12A/CD3 bispecific antibody.
  • the cell expresses two different heavy chains and at least one light chain.
  • the cell expresses a“common light chain” as described herein to reduce the number of different antibody species (combinations of different heavy and light chains).
  • the respective VH regions are doned into expression vectors using methods known in the art for production of bispecific IgG (W02013/157954; incorporated herein by reference), in conjunction with the rearranged human IGKV1 39/IGKJ1 (huVxl 39) light chain.
  • the huVxl 39 was previously shown to be able to pair with more than one heavy chain thereby giving rise to antibodies with diverse specificities, which facilitates the generation of bispedfic molecules (de Kruif, Kramer et al. 2009; W02009/157771).
  • an antibody producing cell that expresses a common light chain and equal amounts of the two heavy chains typically produces 50% bispecific antibody and 25% of each of the monospecific antibodies (i.e. having identical heavy light chain combinations).
  • Several methods have been published to favor the production of the bispecific antibody over the production of the respective monospecific antibodies. Such is typically achieved by modifying the constant region of the heavy chains such that they favor heterodimerization (i.e. dimerization with the heavy chain of the other heavy/light chain combination) over homodimerization.
  • the bispecific antibody of the invention comprises two different immunoglobulin heavy chains with compatible heterodimerization domains.
  • the compatible heterodimerization domains are preferably compatible
  • immunoglobulin heavy chain CH3 heterodimerization domains The art describes various ways in which such hetero-dimerization of heavy chains can be achieved.
  • One preferred method for producing the CLEC12AZCD3 bispecific antibody is disclosed in US 9,248,181 and US 9,358,286.
  • preferred mutations to produce essentially only bispecific full length IgG molecules are the amino acid substitutions L351K and T366K (EU numbering) in the first CH3 domain (the ⁇ K- variant’ heavy chain) and the amino acid substitutions L351D and L368E in the second domain (the‘DE-variant’ heavy chain), or vice versa.
  • the DE-variant and KK-variant preferentially pair to form heterodimers (so-called‘DEKK bispecific molecules). Homodimerization of DE-variant heavy chains (DEDE homodimers) or KK-variant heavy chains (KKKK homodimers) hardly occurs due to strong repulsion between the charged residues in the CH3- CH3 interface between identical heavy chains.
  • the heavy chain/light chain combination that comprises the variable domain that binds CLEC12A comprises a DE variant of the heavy chain.
  • the heavy chain/light chain combination that comprises the variable domain that binds CD3 comprises a KK variant of the heavy chain.
  • binding to membrane-expressed CD3 on HPB-ALL cells can be assessed by flow cytometry (according to the FACS procedure as previously described in W02014/051433).
  • the binding of a candidate CLEC12A/CD3 bispecific antibody to CDS on HPB ALL cells is demonstrated by flow cytometry, performed according to standard procedures known in the art. Binding to cell expressed CD3 is confirmed using CHO cell transfected with CD38/e or CD3y/e.
  • the binding of the candidate bispecific IgGl to CLEC12A is determined using CHO cells transfected with a CLEC12A expression construct; a CD3 monospecific antibody and a CLEC12A monospecific antibody, as well as an irrelevant IgGl isotype control mAb are included in the assay as controls (e.g., an antibody which binds CDS and. another antigen such as tetanus toxin
  • the affinities of the CD3 and CLEC12A Fabs of a candidate CLEC12A/CD3 bispecific antibody for their targets can be measured by surface plasmon resonance (SPR) technology using a BIAcore T100. Briefly, an anti-human IgG mouse monoclonal antibody (Becton and Dickinson, cat. Nr. 556784) is coupled to the surfaces of a CMS sensor chip using free amine chemistry (NHS/EDC). Then the bsAb is captured onto the sensor surface. Subsequently, the recombinant purified antigens human CLEC12A (Sino Biological Inc, cat. Nr.
  • 11896-H07H and human CD36c-Fc protein are run over the sensor surface in a concentration range to measure on- and off-rates. After each cycle, the sensor surface is regenerated by a pulse of HC1 and the bsAb is captured again. Prom the obtained sensorgrams, on- and off-rates and affinity values for binding to human CD3 and CLEC12A are determined using the BIAevaluation software, as previously described (e.g., US 2016/0368988).
  • the T-cell stimulatory capacity of a CLEC12A/CD3 bispecific antibody can be determined in an assay using healthy donor resting T-cells obtained according to the procedure described in W02014/051433 and US 2016/0368988.
  • a candidate CLEC12A/CD3 bispecific antibody is tested in purified healthy donor resting T cells incubated with cells from the leukemia derived HL60 cell line in 10% fetal bovine serum (FBS) or 10% normal human serum (HS) at an effector: target cell ratio of 10:1 or 5:1 for two days. Results are expressed as the percentage of CD69 positive or CD25 positive cells within the CD4 positive or CDS positive T cell population.
  • the ability of a CLEC12A/CD3 bispecific antibody to induce target cell lysis may be tested in an assay using leukemia cells, e.g., HL-60 cells, labeled with carboxyfluorescein diacetate succimidyl ester (CFSE) and co-cultured with T -cells from healthy donor, according to the procedure described in WO2014/051433 and US 2016/0368988.
  • leukemia cells e.g., HL-60 cells
  • CFSE carboxyfluorescein diacetate succimidyl ester
  • a pharmaceutical composition comprising a CLEC12A/CD3 bispecific antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a government regulatory agency or listed in the U.S. Pharmacopeia or another generally recognized pharmacopeia for use in animals, particularly in humans, and includes any and all solvents, salts, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
  • pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, glycerol polyethylene glycol ricinoleate, and the like.
  • Water or aqueous solution saline and aqueous dextrose and glycerol solutions may be employed as carriers, particularly for injectable solutions.
  • Liquid compositions for parenteral administration can be formulated for administration by injection or continuous infusion. Routes of administration by injection or infusion include intravesical, intratumoral, intravenous, intraperitoneal, intramuscular, intrathecal and subcutaneous.
  • the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • compositions suitable for administration to human patients are typically formulated for parenteral administration, e.g., in a liquid carrier, or suitable for reconstitution into liquid solution or suspension for intravenous administration.
  • the compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • solid preparations which are intended for conversion, shortly before use, to liquid preparations for either oral or parenteral
  • Such liquid forms include solutions, suspensions and emulsions.
  • compositions and methods provided herein are particularly useful for activating a T cell or T cells in a patient.
  • the patient has a cancer or is at risk of having cancer.
  • the latter being patients in which are in remission of a cancer and are at risk of recurrence, relapse or metastasis of the cancer.
  • the compositions and methods may be used in the treatment of various myeloid malignancies, including myeloid leukemias and pre-leukemic diseases of myeloid origin.
  • disease that can be treated according to the methods provided herein include myeloid leukemias (e.g., AML and CML) or pre-leukemic diseases such myelodysplastic syndrome (MDS) (and progression to AML), myelofibrosis (MF) or myeloproliferative neoplasm blast phase (MPN-BP).
  • myeloid leukemias e.g., AML and CML
  • pre-leukemic diseases such myelodysplastic syndrome (MDS) (and progression to AML), myelofibrosis (MF) or myeloproliferative neoplasm blast phase (MPN-BP).
  • MDS myelodysplastic syndrome
  • MF myelofibrosis
  • MPN-BP myeloproliferative neoplasm blast phase
  • the bispecific antibody CLEC12A/CD3 bispecific antibody may be combined with another therapeutic molecule. Such combined
  • a CLEC12A/CD3 bispecific antibody may be used in a method for activating a T cell in a subject, wherein the CLEC12A/CD3 bispecific antibody is administered simultaneously, separately or sequentially with an IL-15 moiety.
  • a CLEC12A/CD3 bispecific antibody may be used in the treatment of a cancer in a subject, wherein the CLEC12A/CD3 bispecific antibody may be administered simultaneously, separately or sequentially with an IL-15 moiety.
  • a CLEC12A/CD3 bispecific antibody may be for use in the manufacture of a medicament for activating a T cell in a subject, wherein the CLEC12A/CD3 bispecific antibody is administered simultaneously, separately or sequentially with an IL-15 moiety.
  • a product comprising a CLEC12A/CD3 bispecific antibody and an IL-15 moiety may be a combined preparation for simultaneous, separate or sequential use in activating a T cell in a subject.
  • a CLEC12A/CD3 bispecific antibody may be for use in the
  • a product comprising a CLEC12A/CD3 bispecific antibody and an IL-16 moiety may be a combined preparation for simultaneous, separate or sequential use in treating a cancer in a subject.
  • the IL-10 moiety can be administered according to a suitable dosage, route (e.g., intravenous, intraperitoneal, intramuscular, intrathecal or subcutaneous).
  • a suitable dosage, route e.g., intravenous, intraperitoneal, intramuscular, intrathecal or subcutaneous.
  • CLEC12A/CD3 bispecific antibody and IL-15 moiety can be any suitable CLEC12A/CD3 bispecific antibody and IL-15 moiety.
  • CLEC12A/CD3 bispecific antibody and IL-15 moiety can be formulated for separate administration, wherein they are administered concurrently or sequentially.
  • the IL-Id moiety and the CLEC12A/CD3 bispecific antibody are administered simultaneously.
  • a subject 1 ⁇ 2 administered a single dose of an IL-15 moiety and a single dose of the CLEC12A/CD3 bispecific antibody will be administered repeatedly, over a course of treatment.
  • multiple (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) doses of an IL-15 moiety and multiple (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) doses of a CLEC12A/CD3 bispecific antibody are administered to a subject in need of treatment.
  • CLEC12A/CD3 bispecific antibody may be done weekly, in which regimen, they may be administered on the same day (e.g., simultaneously), or one after the other (e.g., one or more minutes, hours or days before or after one another).
  • the CLEC12A/CD3 bispecific antibody and IL-15 moiety may be, but are not necessarily administered according to the same administration (Le., dosing) protocol.
  • a therapeutically effective dose of the IL-15 moiety may be administered either more or less frequently the CLEC12A/CD3 bispecific antibody.
  • administration of each dose of the IL-15 moiety and the CLEC12A/CD3 bispecific antibody may be on the same day, or alternatively, the IL-15 moiety may be administered 1 or more days before or after the
  • the total daily dosage may be divided and administered in portions during the day if desired. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
  • the IL-15 moiety used in the methods provided herein is recombinant human IL-15 (rhIL-15).
  • the rhIL-15 is administered at doses of about 0.125 pg/kg per day to 2.0 pg/kg per day.
  • the rhIL-15 is administered at 0.125, 0.25, 0.6, 1.0 or 2.0 pg/kg per day.
  • the rhIL-15 is administered by intravenous bolus.
  • the rhIL-15 is administered by continuous intravenous infusion (CIV).
  • CIV intravenous infusion
  • the rhIL-15 is administered by CIV for between 2 to 10 days, 5 to 10 days, or 7 to 10 days.
  • the rhIL-15 is administered by CIV for 10 days.
  • the rhlL- 15 is administered in treatment cycle of between about 30 and 60 days, in which the rhIL-15 is administered by CIV for the first 5 to 10 days of each cycle.
  • the IL-15 moiety administered according to the methods provided herein is a soluble IL-15 receptor or receptor fragment (sIL- 15Ra).
  • the IL-15 moiety is a complex comprising IL-15 and slL- 15Ra, for example, as described in US 9,255,141 and US 9,328,159.
  • the IL-15/IL-15Ra complex is administered at a dose of
  • the IL-15 moiety is an IL-15/IL-15Ra which is administered subcutaneously.
  • the lL-16/lL-16Ra complex is administered at a frequency of every day, every other day, every 3, 4, 5, 6 or 7 days.
  • the IL-15/IL-15Ra is administered 1, 2, 3, 4, 5, 6 or 7 days per week.
  • the dose of the first cycle and each subsequent cycle is 0.1 pg/kg to 1 pg/kg, 1 pg/kg to 5 pg/kg, or 5 pg/kg to 10 pg/kg.
  • the first dose of the first cycle and each subsequent cyde is 0.1 pg/kg to 0.5 pg/kg, 1 pg/kg to 2 pg/kg, 1 pg/kg to 3 pg/kg, 2 pg/kg to 5 pg/kg, or 2 pg/kg to 4 pg/kg.
  • the dose of the first cycle and each subsequent cycle is 0.1 pg/kg, 0.25 pg/kg, 0.5 pg/kg, 1 pg/kg, 1.25 pg/kg, 1.5 pg/kg, 1.75 pg/kg, 2 pg/kg, 2.25 pg/kg, 2.5 pg/kg, 2.75 pg/kg, 3 pg/kg, 3.25 pg/kg, 3.5 pg/kg, 4 pg/kg, 4.25 pg/kg, 4.5 pg/kg, 4.75 pg/kg, or 5 pg/kg.
  • the IL-15/IL-15Ra complex is administered according to a 28 day cycle in which the IL-15/IL-15Ea complex is administered subcutaneously three time per week for two consecutive weeks.
  • the IL-15 moiety used in the methods provided herein is a long-acting form of IL-15, for example, a conjugate of IL-15 and a water soluble polymer as described, for example, in WO 2015815373.
  • IL-15 IL-15 receptor
  • One of ordinary skill in the art can determine the quantity of a long-acting, IL-15 agonist that is sufficient to provide clinically relevant agonist activity at the IL-15 receptor (”IL- 15R").
  • the IL-15 polymer conjugate is administered at a dose of from about 0.001 mg/kg to about 10 mg/kg, preferably from about 0.001 to about 5 mg/kg.
  • the IL-15 polymer conjugate is
  • the IL-15 polymer conjugate is administered at a dose of about 0.03 mg/kg, about 0.1 mg/kg, about 0.3 mg/kg or about 3.0 mg/kg. In other words, the IL-15 polymer conjugate is administered at a dose of about 0.03 mg/kg, about 0.1 mg/kg, about 0.3 mg/kg or about 3.0 mg/kg. In other words, the IL-15 polymer conjugate is administered at a dose of about 0.03 mg/kg, about 0.1 mg/kg, about 0.3 mg/kg or about 3.0 mg/kg.
  • the IL-15 polymer conjugate is administered at a dose of about 0.0025 mg/kg, about 0.008 mg/kg, about 0.01 mg/kg, about 0.025 mg/kg, 0.05 mg/kg or about 0.25 mg/kg.
  • Non-limiting parameters that indicate the treatment method is effective may include one or more of the following: decrease in tumor cells; inhibition of tumor cell proliferation; tumor cell elimination; progression-free survival; appropriate response by a suitable tumor marker (if applicable); increased number of NK (natural killer) cells; increased number of CLEC12A specific T cells; and increased number of CLEC12A specific memory T cells.
  • the frequency of administering the CLEC12AZCD3 bispecific antibody With regard to the frequency of administering the CLEC12AZCD3 bispecific antibody, one of ordinary skill in the art will be able to determine an appropriate frequency. For example, a clinician can decide to administer the CLEC12A/CD3 bispecific antibody relatively infrequently (e.g., once every two weeks) and progressively shorten the period between doses as tolerated by the patient. When an IL-15 moiety is administered, the frequency for these agents can be determined in a similar fashion.
  • Exemplary lengths of time associated with the course of therapy in accordance with the claimed method include: about one week; two weeks; about three weeks; about four weeks; about five weeks; about six weeks; about seven weeks; about eight weeks; about nine weeks; about ten weeks; about eleven weeks; about twelve weeks; about thirteen weeks; about fourteen weeks; about fifteen weeks; about sixteen weeks; about seventeen weeks; about eighteen weeks; about nineteen weeks; about twenty weeks; about twenty -one weeks; about twenty-two weeks; about twenty-three weeks; about twenty four weeks; about seven months; about eight months; about nine months; about ten months; about eleven months; about twelve months; about thirteen months; about fourteen months; about fifteen months; about sixteen months; about seventeen months; about eighteen months; about nineteen months; about twenty months; about twenty one months; about twenty -two months; about twenty -three months; about twenty -four months; about thirty months; about three years; about four years; about five years; perpetual (e.g., ongoing maintenance therapy).
  • the foregoing duration may be associated with one or
  • the efficacy of the treatment methods provided herein can be assessed using any suitable means.
  • the clinical efficacy of the combination treatment is analyzed using AML blast reduction in the bone marrow as an objective response criterion.
  • Patients, e.g., humans, treated according to the methods disclosed herein preferably experience improvement in at least one sign of cancer.
  • one or more of the following can occur: the number of cancer cells can be reduced; cancer recurrence is prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.
  • in vitro assays to determine the T cell mediated target cell lysis (as described in WO2017/010874) can be performed with AML tumor blasts isolated from the subject.
  • the tumor cells are no longer detectable following treatment as described herein.
  • a subject is in partial or full remission.
  • a subject has an increased overall survival, median survival rate, and/or progression free survival.
  • the treatments of the present invention e.g., CLEC12A/CD3 bispecific antibody
  • the combination treatment with an IL-15 moiety may also be used in conjunction with other well-known therapies that are selected for their particular usefulness against the cancer that is being treated.
  • Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when appropriate.
  • chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemotherapeutic agents is described in the Physicians' Desk Reference (PDR), e.g., 1996 edition (Medical Economics Company, Montvale, N.J. 07646-1742, USA); the disclosure of which 1 ⁇ 2 incorporated herein by reference thereto.
  • PDR Physicians' Desk Reference
  • the administration of the chemotherapeutic agent(s) and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent(s) and/or radiation therapy on that disease.
  • the therapeutic protocols e.g., dosage amounts and times of administration
  • the therapeutic protocols can be varied in view of the observed effects of the administered therapeutic agents on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
  • Flow cytometry was performed using a Coidter FC500, Cyan-ADP, Gallios or NAVIOSTM flow cytometer (Beckman Coulter) or FACS CantoA (BD
  • Blasts were defined as SSC+/- CD45DIM and/or CD45POS cells excluding lymphocytes based on CD45/SSC and CD3 (for T cells).
  • T cell activation in cytotoxicity assay with HL60 cells was determined using CD25-PE, 7AAD, CD3-PECY7, CD69-APC, CD8-AF700 and CD4-BV421.
  • Monocyte cytotoxicity was quantified using CD14-FITC, CD16-PE, CD19- ECD, 7AAD, CD3-PECY7, CD69-APC and CD8-AF700.
  • T cells were visualized with 7AAD, CD3- PECY7, CD8-AF700 and CD4-BV421.
  • Cytotoxicity of CD34POS bone marrow progenitor cells was quantified using a) CLEC12A-PE (Biolegend), 7AAD, CD34-PECY7, CD10-APC-AF760 and CD5- BV421; and b) with CD90-FITC, CLEC12A-PE (Biolegend)/IgG2a-PE, CD38-ECD, 7AAD, CD34-PECY7, CD10-APC, CD45RA-AF700, CD123-BV421 or CD135a- BV421 and Lineage (CD3, CD14, CD16, CD19, CD20, CD56)-BV510.
  • CLEC12A expression on AML blast» used for cytotoxicity assays with AML blasts and purified T cells was quantified using a) CLEC12A-PE (R&D Systems), CD14-ECD, CD33-PECY5, CD38-PECY7, CD117-APC, CD19-AF700, CD34-BV421 and CD45-krome orange; or b) CLEC12A-PE (R&D Systems), CD19-ECD, CD3- PECYo, CD66-PECY7 (Biolegend), CD8-AF700, CD4-BV421 and CD4o-krome orange.
  • CLEC12A expression on AML blasts was determined using a) (1,000 ng/mL condition) CD14-FITC, CLEC12A-PE (R&D Systems)ZIgG2b-PE CD34-ECD, 7AAD, CD33-PECY7, CD8-AF700, CD4-BV421 and CD45-krome orange; or b) (200 ng/mL condition) CD14-F1TC, CLEC12A-PE (R&D Systems), CD34-ECD, CD4- PECY5.5, CD117-APC, CD8-AF700, CD33-APC AF750 and CD45-krome orange.
  • Efficacy of antibody MF4327xMF5196 in primary AML blast samples was assessed using a) (1,000 ng/mL condition) CD14-FITC, CD34-ECD, 7AAD, CD33- PECY7, CD3-APC, CD8-AF700, CD4-BV421 and CD45-krome orange; or b) (200 ng/mL condition) CD3-FITC, 7AAD, CD4-PECY7, CD8-AF647, CD45RA-AF700, CD33-APC AF750 and CD45-krome orange.
  • Antibody MF4327xMF5196 as used in the examples and the figures referred to in the examples is a full length human lgGl bispecific antibody specific for CLEC12A and CD3 with a common light chain variable region of IgVkl-39*01 of SEQ ID: NO 56 in the sequence listing summary. It has the heavy chain variable region of MF4327 and the heavy chain variable region of MF5196 depicted in the sequence listing summary. The constant part of the antibody can be effector function silenced or not as indicated.
  • the negative control antibodies were a cLC monospecific antibody against tetanus toxoid (TTxTT IgG, isotype control (de Kruif, Kramer et al. 2009)) and a cLC bispecific antibody against tetanus toxoid and CDS (MockxCDS or TTxCD3 IgG, using the same CD3 Fab as in antibody
  • the positive control antibody was a bivalent cLC monospecific antibody against CD3 (CD3xCD3, also using the same CDS Fab as in antibody MF4327xMF5196).
  • bispecific antibodies electrostatically engineered heavy chains were used for transient production, resulting in bispecific IgGl with >95% purity (Gunasekaran, Pentony et al. 2010). Later batches were produced using heavy chain Fc engineering wherein proprietary mutations involving charged residues at the CH3 interface were introduced (De Nardis, Hendriks et al. 2017). Unless otherwise specified, the IgG batches of antibody MF4327xMF5196, MockxCDS and TTxTT IgG lacked Fc effector function upon introduction of changes in the CH2 regions.
  • Antibody MF4327xMF5196 was immobilized on a CMS chip (GE).
  • C57B1/6J mice received a single intravenous dose of 1 mg/kg antibody MF4327xMF5196.
  • Antibody MF4327xMF5196 serum levels were quantified at various time points using an anti-human IgG ELISA (ZeptoMetrix).
  • HL60 cells were cultured in IMDM (GIBCO Invitrogen, 21980-065) containing 2mM 1-glutamine and 10% FBS (Integra).
  • IMDM Gibco Invitrogen, 21980-065
  • FBS Integra
  • CHO-K1 DSMZ
  • Jurkat E6.1 LGC
  • J.RT-T3.5 LGC
  • ThermoFisher Scientific ThermoFisher Scientific.
  • the CLEC12A-CHO-K1 cell line was generated by stable integration of pcDNA3.1+ vector (ThermoFisher Scientific) encoding human CLEC12A.
  • AML patient samples (Table IP) and healthy donor bone marrow were obtained in accordance with the Declaration of Helsinki and RUMC institutional guidelines and regulations.
  • IMDM medium containing 7% dimethyl sulfoxide (DMSO; Sigma-Aldrieh, W387509) and 10% foetal bovine serum (FBS, Integra).
  • FBS foetal bovine serum
  • HS human serum
  • gg/mL DNAse Roche, 11284932001
  • MgCla 1.25 mM MgCla
  • PBMCs Human peripheral blood mononuclear cells
  • PB peripheral blood
  • CDS 1*08 T cells were purified from freshly isolated or cryopreserved PBMCs by negative selection by using magnetic cell sorting (MACS, Miltenyi, 130-096-535). Cytotoxicity assays were performed in IMDM supplemented with 10% AB+ HS (Sanquin) in 96-well plates (NuncTM, ThermoFisher Scientific). Unless otherwise specified, 10 s T cells were used. CFSE-labelled HL60 or primary AML blast target cells were used (ThermoFisher Scientific, Cl 1-57).
  • AML blasts Prior to CFSE labelling, AML blasts were precultured overnight in assay medium supplemented with 100 ng/mL GM-CSF (tmmunotools, 11343125), 100 ng/mL G-CSF (Amgen Inc, Neupogen®), 50 ng/mL IL-3 (Cellgenix), 25 ng/mL SCF (Immunotools, 11343325) and 20 ng/mL Flt3L (Immunotools, 11343305). Subsequently, T cells and target cells were co -cultured in the same cytokine-supplemented assay medium in the presence of antibody MF4327xMF5196 or control IgGs.
  • PBMCs (2x10 s ) were incubated in IMDM with 10% AB+ HS for 48 hours in the presence of antibody MF4327xMF5196 or control IgGs. Prior to harvesting, the assay plates were centrifuged (500g, 5 minutes) to collect supernatant for cytokine quantification. After a subsequent incubation at 2-8°C for 1 hour, cells were stained in the assay medium without washing. The specific lysis of monocytes, B cells and natural killer (NK) cells was quantified by flow cytometry in a fixed volume collected from each assay well (Fig 8).
  • CFSE-labelled CD3 POS T cells were co-cultured with CD14 POS MACS- purified (Miltenyi, 130-050-201) autologous monocytes in IMDM with 10%AB+ HS with or without test IgG at an E:T ratio of 5: 1. After 5 days, the fraction of proliferating T cells was visualized as CFSE ⁇ w T cells by flow cytometry.
  • IL-lB The levels of IL-lB, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, GM-CSF, TNF-a and IFN-g in culture supernatants were measured using the Luminex® platform (Life Technologies, LHCOOOl).
  • CDS 1*08 T cells and CD34 pos cells were isolated from healthy donor bone marrow mononuclear cells (obtained from orthopaedic patients or purchased from Lonza, 2M-125C) by positive selection using MACS (Miltenyi, 130-046-702 and 130-050-101, respectively).
  • Pre-activated T cells were generated by activating purified T cells for 6 days in plates pre-coated with anti-CD3 (1 pg/mL, BD
  • CFU colony forming unit
  • AML patient samples (5xl0 5 cells) were cultured in the presence of antibody MF4327xMF5196 or control bispecific IgG in IMDM containing 10% AB+ HS, 2.5 ng/mL GM-CSF, 12.5 ng/mL G-CSF (Neupogen, Amgen), 6.26 ng/mL IL-3 (Immunotools, 11340035), 3.0 ng/mL SCF and 2.5 ng/mL Flt3L After 24 hours, 20 ng/mL IL-15 (Miltenyi, 130-095-766) was added to the culture. After 10 days, AML blast lysis and T cell expansion was quantified by flow cytometry.
  • Antibody MF4327xMF 5196 dosing regimen in human AML patients is provided.
  • the MF4327xMF5196 antibody is being explored as a single agent in a first in human study using a dose escalation in cohorts of 3-6 patients.
  • Patients received the antibody with a regimen of administration designed to provide an efficacious and safe dose.
  • the antibody was administered in infusions of 2-4 hours.
  • the treatment was planned with time intervals.
  • the regimen of administration was designed to prevent and minimize the severity and incidence of Cytokine release syndrome (CRS).
  • the treatment regimen has components that are applied at the first cycle (during the first 28 days).
  • the priming dose (ClDl); a step-up and fixed full dosing (C1D4, C1D8, C1D15) and the use of pre- medication.
  • a priming dose the first administration (ClDl) is given with a low dose (1 mg, 3 mg, 5 mg).
  • Step-up dosing subsequent doses after ClDl are given with gradual increments of the dose until the planned full dose is given.
  • the step-up dosing includes more frequent administration during the first week. In the first week the treatment is given every 3 days (D1-D4-D8) and then the treatment is given every 7 days (on weekly basis).
  • patients received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 15 mg on day 8, and a full dose of 25 mg on day
  • patients received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15.
  • patients received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15.
  • the patient’s response to the treatment was followed, including analysis of cytokine levels before each dose, and 4 hours and 24 hours after the end of infusion.
  • Antibody MF4327xMF5196 binds specifically to CLEC12A pos and CD3 POS cells within the normal hemopoietic compartment
  • Antibody MF4327xMF5196 is a T cell-targeting bispecific antibody that has been developed to treat CLECl2a positive cancer cells, such as present in AML. It binds to CD3 on T cells and CLEC12A on the AML blasts and LSCs.
  • Antibody MF4327xMF5196 is a human full-length IgGl bispecific antibody based on a common light chain (cLC) bispecific IgG format.
  • CLEC12A but not to parental CHO-Kl cells. Via its CD3 arm, it bound to CDS ⁇ 8 JurkatE6.1 cells, but not to CD3 NKG Jurkat J.RT-T3.5 cells.
  • the affinity of antibody MF4327xMF5196 for its target proteins was 3 nM for CLEC12A and 177 nM for CD3, as determined by surface plasmon resonance technology using recombinant proteins.
  • PK pharmacokinetics
  • antibody MF4327xMF3196 also bound to monocytes, myeloid dendritic cells (mDC), plasmacytoid dendritic cells (pDC) and granulocytes, but not to natural killer (NK) cells or B cells (Fig IB and data not shown).
  • mDC myeloid dendritic cells
  • pDC plasmacytoid dendritic cells
  • NK natural killer cells or B cells
  • Antibody MF4327xMF5196 bound uniformly to granulocyte-macrophage progenitor (GMF) cells, whereas it bound only a minor fraction of the common myeloid progenitor (CMP) and megakaryocyte- erythroid progenitor (MEF) subsets. Notably, antibody MF4327xMF5196 binding was not observed within the CD34 pos CD38 Nm compartment, which includes the multipoient progenitors (MPP) and pluripotent hemopoietic stem cells (HSC).
  • MMF common myeloid progenitor
  • MEF multipoient progenitors
  • HSC pluripotent hemopoietic stem cells
  • Antibody MF4327xMF 5196 induces CLECl 2A-specific T cell activation and lysis of CLECl 2A pos HL60 cells
  • CLECl2A pos target cells in a cytotoxicity assay.
  • the effector cells were healthy donor-derived resting T cells and the target cells were CLEC12A P0S HL60 cells.
  • an MF4327xMF5196 bispecific antibody with a wild- type (WT) Fc region (intact Fc effector function) was compared with MockxCD3 control IgG formatted with a WT Fc region.
  • WT wild- type
  • the MF4327xMF5196 bispecific antibody induced upregulation of CD69 (Fig 2A) and CD25 (data not shown) on CD4 and CDS T cells after 24 and 48 hours.
  • T cell-mediated lysis of HL60 cells was already evident after 24 hours and >95% HL60 cell cytotoxicity was measured after 48 hours (Fig 2B).
  • Target cell lysis correlates to the effector-to-target ratio (E:T ratio) with a maximum lysis at E:T ratios of 5:1 or higher (Fig 9).
  • the CLECl2AxCD3- induced lytic activity was CLEC12A antigen-mediated (Fig 2 A, B); only at later time points some T cell activation and HL60 target cell lysis was observed for the MockxCD3 control. Nevertheless, this revealed that the bispecific IgG format with WT Fc could induce Fey receptor-mediated T cell activation without the need for CLEC12A binding (Fig 2A and B, 72 hours).
  • Antibody MF4327xMF5196 with silenced Fc effector function induces specific lysis of CLEC12ATM AML cell line
  • a bispecific IgG format lacking Fc effector function was generated by introducing changes into the CH2 region of both IgG l heavy chains.
  • a variety of means of mitigating Fc effector function are known in the art. Binding to CD16, CD32 and Clq, and diminished binding to CD64 >200-fold was accomplished via Fc engineering modifying residues in the CH2 domain.
  • antibody MF , 4327xMF , 5196 induced upregulation of CD69 (Fig 2C) and CD25 (Fig 2D) on CD4 T cells and CDS T cells.
  • Antibody MF4327xMF5196 induces specific lysis of CLEC12A FOS monocytes by autologous T cells
  • the capacity of antibody MF4327xMF5196 to induce redirected lysis of CLEC12A P0S monocytes by resting autologous T cells was determined in PBMC cultures.
  • Antibody MF4327xMF5196 induced activation of CD4 T cells and CDS T cells (Fig 3A), and lysis of monocytes (from 100 ng/mL, Fig 3B).
  • the antibody MF4327xMF5196-induced activity was CLECl2A-specific as the
  • MockxCDS control did not induce observable T cell activation or monocyte lysis at concentrations up to 10,000 ng/mL (Fig 3A-B). Furthermore, the lytic activity of antibody MF4327xMF5196-activated cytotoxic T cells was selective for CLEC12A- antigen expressing cells, as B cell and NK cell fractions were not affected, even at the highest tested antibody MF4327xMF5196 concentrations (Fig 3C and data not shown).
  • Antibody MF4327xMF5196 induced the release of IL-lB, TNF-a, IFN-g, IL-6 and IL-10 in the range of 10-200 pg/mL, and IL-8 release in the range of 10,000-30,000 pg/mL (Fig 3E).
  • antibody MF4327xMF5196-induced cytokine levels were lower than those obtained after stimulation with an anti-CD3 bivalent monospecific antibody.
  • the antibody MF4327xMF5196-induced cytokine release was CLEC12A-specific as cytokine release in the presence of the MockxCDS control was absent or limited.
  • Antibody MF4327xMF5196-induced targeting of normal CD 34+ cells spares erythrocyte and megakaryocyte differentiation as well as retains potential to develop the mono-myelocytic lineage
  • MF4327xMF5196 a subset that comprises CMPs (Fig 4C).
  • MF4327xMF5196- induced redirected lysis was evaluated at a functional level using CFU assays.
  • the outgrowth of the erythroid (BFU-E; burst-forming units- erythroid) or megakaryocyte (CFU-Mk; colony forming unit-megakaryocyte) colonies from the bone marrow CD34 pos hemopoietic progenitor cells were not affected by antibody MF4327xMF5196 (Fig 4D).
  • antibody MF4327xMF5196 induced redirected lysis caused a strong reduction in GMPs only a modest reduction of 24% in the outgrowth of granulocyte-macrophage colonies (CFU-GM) was observed at functional level.
  • GM-progenitor cells unaffected by antibody MF4327xMF5196, retained their capacity to develop both CD14 POS CD36 pos CD33 m8 monocytic and CDIS ⁇ DSS 1108 myelocytic cells (Fig 4E), as determined by flow cytometric analysis of the CFU-GM colonies.
  • Antibody MF4327xMF5196 induces redirected lysis of primary AML blasts by autologous T cells
  • CM central memory
  • EM effector memory
  • EMRA+ reacquired CD45RA
  • the capacity of antibody MF4327xMF5196 to induce CLKCl2A-specific lysis of primary AML blasts by resting autologous T cells collected from the same AML patient at a later time point was determined (Fig 5A, patients listed in Table III, patients 7-9).
  • the AML blasts were cocultured with autologous resting T cells taken during clinical remission at an E:T ratio of 5:1 for 48 hours.
  • Antibody MF4327xMF5196 induced potent CLEC12A- spedfic activation of CD4 and CDS T cells as well as >70% AML blast lysis (Fig 5C- E).
  • Antibody MF4327xMF5196 redirects lysis of primary AML blasts in diagnostic samples with low E:T ratios
  • MF4327xMF5196 was 41-1591%. Antibody MF4327xMF5196 and MockxCD3 control IgG were added once at the onset of culture. T cell expansion and AML blast lysis was quantified by flow cytometry at day 7 and day 10. Not only did antibody MF4327xMF5196 efficiently induce CLEC 12A- mediated T cell expansion (range of fold expansion 6-157), it also induced AML blast lysis at day 7 and day 10 (Fig 6). At day 10, antibody MF4327xMF5196 had efficiently induced AML blast lysis (23-98%) in 10/11 patient samples. Although the MockxCD3 control antibody also reduced the numbers of tumor cells in some samples, in those samples the levels of T cell activation were much lower than those observed for antibody MF4327xMF5196 (median of 48 vs 2-fold T cell expansion by antibody
  • MF4327xMF5196 vs MockxCD3.
  • antibody MF4327xMF5196 was active even in AML samples with very low effector-to-target ratios (Fig 6 and Table P).
  • Fig 6 and Table P effector-to-target ratios
  • antibody MF4327xMF5196 efficiently induced CLECl2A-mediated lysis of AML blasts by T cells present in AML patient bone marrow samples, even at very low E:T ratios, and also provoked robust T cell proliferation.
  • Figure 14 shows the data obtained from patients that received a priming dose of 1 mg on day 1, followed by step-up doses of 3 mg on day 4 and 16 mg on day 8, and a full dose of 26 mg on day 15.
  • Figure 15 shows the data obtained from patients that received a priming dose of 3 mg on day 1, followed by step-up doses of 10 mg on day 4 and 25 mg on day 8, and a full dose of 40 mg on day 15.
  • Figure 16 shows the data obtained from patients that received a priming dose of 5 mg on day 1, followed by step-up doses of 15 mg on day 4 and 25 mg on day 8, and a full dose of 60 mg on day 15..
  • Antibody MF4327xMF5196 as referred to in the examples is a full-length antibody.
  • the Fc effector silenced version of the CLECl2AxCD3 bispecific human IgGl antibody is shown to be able to treat all subtypes of AML by targeting CLEC12A on AML blasts and leukemic stem cells (LSCs).
  • Antibody MF4327xMF5196 as referred to in the examples is a full-length antibody.
  • the Fc effector silenced version of the CLECl2AxCD3 bispecific human IgGl antibody is shown to be able to treat all subtypes of AML by targeting CLEC12A on AML blasts and leukemic stem cells (LSCs).
  • MF4327xMF5196 is a potent bispecific antibody that efficiently activates and redirects T cells to lyse CLECl2A-expressing cells such as AML cells.
  • this CLEC12AxCD3 T cell engager efficiently induced CLECl2A-mediated lysis of AML blasts by redirecting resident autologous T cells.
  • AML samples were cultured for 7-10 days in the presence of antibody MF4327xMF5196. This assay establishes antibody MF4327xMF6196 efficacy in two ways: it revealed that antibody
  • MF4327xMF5196 had the capacity to induce expansion of AML patient T cells in these samples, and it showed that those antibody MF4327xMF5196-redirected T cells were able to redirect cytolytic activity towards AML blasts through cytolysis (presented in Table II). Antibody MF4327xMF5196 was able to induce significant T cell expansion and blast lysis even in those AML samples that had low initial E:T ratios (1:45-1:97) or relatively low levels of CLEC12A expression.
  • MF4327xMF5196 was observed compared to minimal activity for MockxCD3 in 9/10 primary AML samples.
  • AML patient-derived T cells Although healthy donor T cells were used in many initial experiments, cytotoxicity assays were also performed using AML patient-derived T cells to test the capability of antibody MF4327xMF5196 to redirect T cells in a disease setting. Specific defects have been reported for AML T cells, including impaired
  • AML patient-derived T cells can be functionally activated by a full-length IgG CD3-engaging bispecific IgG, resulting in T cell activation and proliferation, in a HSC -sparing manner and at low E:T ratios.
  • MF4327xMF 5196 effectively enlarges the effector T cell compartment, thus facilitating more favorable E:T ratios for efficient eradication of AML blasts and LSCs in patients.
  • antibody MF4327xMF5196 binding is restricted to the myeloid compartment, as antibody MF4327xMF5196 is capable of a CLEC12A- spedfic targeting, sparing other regions of the bone marrow.
  • antibody MF4327xMF5196 binds in the bone marrow uniformly to GMP cells, while it only binds a minor fraction of the CMP and MEP subsets. More
  • antibody MF4327xMF5196 does not bind to the CD34 pos CD38 Nm compartment which includes pluripotent HSCs. In line with the minor CLEC12A expression on the CMP and MEP fractions it is shown that antibody
  • MF4327xMF5196 does not affect the development of eiythroid nor megakaryocytic lineage in our in vitro CFU assays. Furthermore, although antibody MF4327xMF5196 reduced the number of CLEC12A 1 ’ 08 GMP cells in the ex vivo cytotoxicity assay, it is observed in the follow-up CFU assays that upon antibody MF4327xMF5196 treatment the remaining CD34 ms bone marrow cells were able to give rise to both the monocytic and myelocytic lineages, permitting mono- myelocytic outgrowth arising from CD34 P0S CLEC12A N1BG progenitor cells, including HSCs and MPPs, which are not observably impacted by the treatment of antibody MF4327xMF5196 (van Rhenen, van Dongen et al. 2007, Notta, Zandi et al. 2016, Bill, van Kooten Niekerk et al. 2018).
  • hemopoietic cells after treatment with antibody MF4327xMF5196 contrasts with the likely situation for treatments that target CD33 or CD 123 (Aigner, Feulner et al. 2013, Al-Hussaini, Rettig et al. 2016).
  • CD33 and CD 123 are more widely expressed on normal CD34 P0S progenitors, including CMPs, GMPs and pluripotent HSCs (Taussig, Pearce et al. 2005).
  • the present invention shows that CLEC12A targeting spares the normal CD123 P0S CMPs which are a target for CD 123 directing therapies.
  • CD33 and CD 123 targeting chimeric antigen receptor (CAR)- transduced T cells induce lysis in the CD34 P0S CD38 NE ° stem cell fraction (Pizzitola, Anjos- Afonso et al. 2014, Kenderian, Ruella et al. 2015).
  • CAR chimeric antigen receptor
  • the Fc region of antibody MF4327xMF5196 has been modified to prevent binding to FcyR and Clq, without affecting binding to the FcRn receptor.
  • the results of the monocyte assays indeed showed that antibody MF4327xMF5196- induced T cell activation and associated cytokine release is restricted to CLEC12A expressing cells.
  • the Fc silencing dampens typical antibody dependent cellular cytotoxicity (ADCC) and Clq-mediated complement-dependent cytotoxicity (CDC) of CDS 1108 and/or CLEC12A P0S cells.
  • ADCC antibody dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • MF4327xMF5196-activated T cells lyse CLEC12A P0S cells selectively (e.g. B cells and NK cells were unaffected in PBMC cultures).
  • mice confirmed that antibody MF4327xMF5196, a full-length IgGl, has a normal FcRn- mediated in vivo half-life of 9-10 days. This means that— unlike other T cell engager concepts— effective systemic levels of antibody
  • MF4327xMF5196 in patients can be achieved by weekly intravenous
  • Antibody MF4327xMF5196 selectively targets myeloid blasts while sparing normal HSCs. Based on its potential to eradicate residual LSCs, antibody
  • MF4327xMF5196 is considered for the management of minimal residual disease (MRD) in AML.
  • CD34+ cells were collected from the bone marrow of MPN-BP patients.
  • CD34+ cells were sorted at the basis of forward scatter (FSC), side scatter (SSC), CD45, CD34, CD38 and CLEC12A expression. Cells with low SSC and that were CD45 dim ,
  • CD34*, CD38 were sorted into two fractions on the basis of CLEC12A expression.
  • the cells were plated in MethoCultTM H4435 Enriched (STEMCELL technologies) supplemented with SCF, IL-3, 1L-6, EPO, G-CSF, and GM-CSF and incubated for 12-14 days. Colonies were picked and re-plated in MethoCultTM H4435 Enriched (STEMCELL technologies) supplemented with SCF, IL-3, IL-6, EPO, G-CSF, and GM-CSF and re-incubated for 12-14 days.
  • the results of the first plating were that the CLEC12A negative fraction had 20 big colonies of (40 cells or more) and 10 small colonies ( ⁇ 40 cells per colony) per 1500 plated cells.
  • the CLEC12A positive fraction had no big and 32 small colonies per 1500 plated cells.
  • the colonies in the plates of the CLEC12A negative fraction were not able to generate secondary colonies in the re-plating experiment, zero big or small colonies per 1500 seeded cells.
  • re-plating the cells from the CLEC12A first plating resulted in 44 small colonies per 1500 cells, of these approximately 80% was positive for the JAK2 driver mutation, see Figure 13.
  • CD34posCD38-CLECl2A- cells from MPN-BP patient are only able to generate hemopoietic colonies (CFU-GM) in primary culture, which are
  • CD34-CD38- stem cells in MPN-PB patients expressing CLEC12A are capable of generating blast colonies that can be serially re-plated and therefore contain leukemic initiating capacity.
  • Differential CLEC12A expression can be used to identify, eradicate and/or isolate leukemic initiating cells (LIC) from patients with MPN-BP.
  • LIC leukemic initiating cells
  • a CD3 - CLEC1.2A bispecific antibody as disclosed herein can be used to eradicate these LIC in MPN-PB, to reduce the number of MPN blast phase cells and thereby alleviate the disease in patients with MPN-BP.
  • T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of
  • C-type lectin-like molecule- 1 a novel myeloid cell surface marker associated with acute myeloid leukemia. Cancer Res 64(22): 8443-8450.
  • DCAL-2 Dendritic-cell-associated C-type lectin 2
  • KLRL1 a novel killer cell lectinlike receptor, inhibits natural killer cell
  • TIM-3 as a novel therapeutic target for eradicating acute myelogenous leukemia stem cells. Int J Hematol 98(6): 627- 633.
  • M1CL myeloid inhibitory C-type lectin-like receptor
  • Needleman SB Wunsch CD. A general method applicable to the search for similarities in the amino acid sequence of two proteins. J Mol Biol. 1970;48(3):443- 453.
  • Table I Mean ECso values of antibody MF4327xMF5196-induced T cell activation and target cell lysis
  • Antibody MF4327xMF5196 ECso values were determined in an HL60 cytotoxicity assay. Data are from 6 healthy donors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
EP19835540.6A 2018-12-20 2019-12-20 Clec12axcd3 bispecific antibodies and methods for the treatment of disease Pending EP3898685A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18214478 2018-12-20
PCT/NL2019/050864 WO2020130829A1 (en) 2018-12-20 2019-12-20 Clec12axcd3 bispecific antibodies and methods for the treatment of disease

Publications (1)

Publication Number Publication Date
EP3898685A1 true EP3898685A1 (en) 2021-10-27

Family

ID=65013454

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19835540.6A Pending EP3898685A1 (en) 2018-12-20 2019-12-20 Clec12axcd3 bispecific antibodies and methods for the treatment of disease

Country Status (7)

Country Link
US (1) US11873338B2 (zh)
EP (1) EP3898685A1 (zh)
JP (1) JP2022521036A (zh)
CN (2) CN114344461A (zh)
MA (1) MA54525A (zh)
TW (1) TW202034952A (zh)
WO (1) WO2020130829A1 (zh)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015037311A1 (ja) * 2013-09-10 2015-03-19 日立化成株式会社 スラリー、研磨液セット、研磨液、基体の研磨方法及び基体
JP2022521036A (ja) 2018-12-20 2022-04-05 メルス ナムローゼ フェンノートシャップ CLEC12AxCD3二重特異性抗体及び疾患の治療方法
WO2023002392A1 (en) * 2021-07-20 2023-01-26 Abl Bio Inc. Anti-cll-1/anti-cd3 bispecific antibodies and uses thereof
WO2023164305A1 (en) * 2022-02-28 2023-08-31 Jasper Therapeutics, Inc. Compositions and methods for depletion of diseased hematopoietic stem cells
WO2024036139A2 (en) * 2022-08-09 2024-02-15 Qlsf Biotherapeutics, Inc. Antibodies binding to clec12a

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2526284C (en) 2003-06-25 2014-11-18 Crucell Holland B.V. Binding molecules for the treatment of myeloid cell malignancies
BRPI0511782B8 (pt) 2004-06-03 2021-05-25 Novimmune Sa anticorpos anti-cd3, uso e método de produção dos mesmos, composição farmacêutica, molécula de ácido nucleico isolada e vetor
US10118970B2 (en) 2006-08-30 2018-11-06 Genentech, Inc. Multispecific antibodies
SI3456190T1 (sl) 2008-06-27 2022-06-30 Merus N.V. Transgena mišja živali, ki proizvaja protitelesa
CN104447995A (zh) 2009-03-20 2015-03-25 霍夫曼-拉罗奇有限公司 双特异性抗-her抗体
JP6251570B2 (ja) 2010-09-21 2017-12-20 アルター・バイオサイエンス・コーポレーション 多量体il−15可溶性融合分子並びにその製造及び使用方法
NZ772318A (en) 2012-04-20 2023-06-30 Merus Nv Methods and means for the production of ig-like molecules
JP6471095B2 (ja) 2012-09-27 2019-02-20 メルス ナムローゼ フェンノートシャップ T細胞エンゲージャーとしての二重特異性IgG抗体
EP3028538B1 (de) 2013-08-02 2024-05-22 BSH Hausgeräte GmbH Kochfeldvorrichtung
TW201718647A (zh) * 2015-06-16 2017-06-01 建南德克公司 抗-cll-1抗體及使用方法
WO2017010874A1 (en) * 2015-07-10 2017-01-19 Merus N.V. Human cd3 binding antibody
WO2017167350A1 (en) 2016-03-30 2017-10-05 Horst Lindhofer Multispecific antibodies for use in the treatment of a neoplasm of the urinary tract
JOP20170091B1 (ar) * 2016-04-19 2021-08-17 Amgen Res Munich Gmbh إعطاء تركيبة ثنائية النوعية ترتبط بـ cd33 وcd3 للاستخدام في طريقة لعلاج اللوكيميا النخاعية
CN109923128A (zh) * 2016-11-15 2019-06-21 基因泰克公司 用于用抗cd20/抗cd3双特异性抗体进行治疗的给药
JP2022521036A (ja) 2018-12-20 2022-04-05 メルス ナムローゼ フェンノートシャップ CLEC12AxCD3二重特異性抗体及び疾患の治療方法

Also Published As

Publication number Publication date
CN113366018A (zh) 2021-09-07
MA54525A (fr) 2021-10-27
US20220010013A1 (en) 2022-01-13
JP2022521036A (ja) 2022-04-05
WO2020130829A1 (en) 2020-06-25
US11873338B2 (en) 2024-01-16
CN114344461A (zh) 2022-04-15
TW202034952A (zh) 2020-10-01

Similar Documents

Publication Publication Date Title
AU2018204173B2 (en) Bispecific IgG antibodies as T cell engagers
US11873338B2 (en) CLEC12AxCD3 bispecific antibodies and methods for the treatment of disease
Busfield et al. Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC
EP4275702A2 (en) Cd70 combination therapy
JP2022169504A (ja) 新規な抗SIRPa抗体およびそれらの治療適用
AU2016276509A1 (en) Methods and antibodies for modulation of immunoresponse
CN115698062A (zh) 表达嵌合抗原受体并具有降低的促炎细胞因子信号传导的经基因修饰的免疫细胞
US20200384084A1 (en) Use of bispecific antibody and il-15 for combination therapy
EP4389767A1 (en) Monoclonal antibody targeting sirp? and use thereof
US20210363252A1 (en) Compositions comprising a t cell redirection therapeutic and a vla-4 adhesion pathway inhibitor
WO2024138072A1 (en) Methods of treating autoimmune disorders using multimeric anti-cd38/anti-cd3 antibodies
WO2023023491A1 (en) Anti-flt3 antibodies, cars, car t cells and methods of use
CA3229520A1 (en) Bispecific anti-flt3/cd3 antibodies and methods of use
Griffiths Targeting OX40 with monoclonal antibodies as a form of cancer immunotherapy
EA040393B1 (ru) БИСПЕЦИФИЧЕСКИЕ АНТИТЕЛА КЛАССА IgG В КАЧЕСТВЕ РЕКРУТЕРОВ Т-КЛЕТОК

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210623

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40061757

Country of ref document: HK

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MERUS N.V.

RIN1 Information on inventor provided before grant (corrected)

Inventor name: WASSERMAN, ERNESTO ISAAC

Inventor name: SIRULNIK, LEONARDO ANDRES

Inventor name: VAN LOO, PIETER FOKKO

Inventor name: BOL, CORNELIS JACOB JOHANNES GEORGE

Inventor name: BAKKER, ALEXANDER BERTHOLD HENDRIK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20240409